WO2006080582A1 - Agent for control of function of antigen-presenting cell - Google Patents

Agent for control of function of antigen-presenting cell Download PDF

Info

Publication number
WO2006080582A1
WO2006080582A1 PCT/JP2006/301933 JP2006301933W WO2006080582A1 WO 2006080582 A1 WO2006080582 A1 WO 2006080582A1 JP 2006301933 W JP2006301933 W JP 2006301933W WO 2006080582 A1 WO2006080582 A1 WO 2006080582A1
Authority
WO
WIPO (PCT)
Prior art keywords
zinc ion
cells
function
zinc
antigen
Prior art date
Application number
PCT/JP2006/301933
Other languages
French (fr)
Japanese (ja)
Inventor
Toshio Hirano
Hidemitsu Kitamura
Original Assignee
Riken
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Riken filed Critical Riken
Priority to US11/815,071 priority Critical patent/US20090054316A1/en
Priority to EP06713076A priority patent/EP1854483A4/en
Publication of WO2006080582A1 publication Critical patent/WO2006080582A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • A61K31/197Carboxylic acids, e.g. valproic acid having an amino group the amino and the carboxyl groups being attached to the same acyclic carbon chain, e.g. gamma-aminobutyric acid [GABA], beta-alanine, epsilon-aminocaproic acid, pantothenic acid
    • A61K31/198Alpha-aminoacids, e.g. alanine, edetic acids [EDTA]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4402Non condensed pyridines; Hydrogenated derivatives thereof only substituted in position 2, e.g. pheniramine, bisacodyl
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/47064-Aminoquinolines; 8-Aminoquinolines, e.g. chloroquine, primaquine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • A61K33/30Zinc; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/02Nasal agents, e.g. decongestants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/10Drugs for disorders of the urinary system of the bladder
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/04Antipruritics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/14Decongestants or antiallergics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/12Drugs for disorders of the metabolism for electrolyte homeostasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/14Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/38Drugs for disorders of the endocrine system of the suprarenal hormones
    • A61P5/40Mineralocorticosteroids, e.g. aldosterone; Drugs increasing or potentiating the activity of mineralocorticosteroids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/04Antihaemorrhagics; Procoagulants; Haemostatic agents; Antifibrinolytic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • G01N33/5047Cells of the immune system
    • G01N33/5052Cells of the immune system involving B-cells

Definitions

  • the present invention relates to a drug capable of controlling the function of antigen-presenting cells. More specifically, the present invention relates to a drug capable of controlling an immune response reaction through activation of antigen-presenting cells.
  • Antigen-presenting cells such as dendritic cells are central to the control of the strength and quality of the immune response, and are being used for autoimmune diseases, allergic diseases, cancer immunotherapy, and the like.
  • research on culture methods that control the growth and activation of antigen-presenting cells is underway.
  • Currently, there is still a need for a method that efficiently and specifically controls the function of antigen-presenting cells. is there.
  • An object of the present invention is to provide a novel function-controlling agent for antigen-presenting cells. Specifically, the present invention controls immune responses via activation of antigen-presenting cells.
  • the purpose is to provide a drug capable of Furthermore, the present invention applies the drug to various diseases such as treatment of autoimmune diseases, treatment of allergic diseases, suppression of rejection during organ transplantation, and further development of a method for enhancing immune response against cancer, The purpose is to develop an efficient vaccine.
  • the present inventors have found that the function of antigen-presenting cells such as dendritic cells can be controlled by controlling the zinc ion level. Furthermore, it was confirmed that the function control of the antigen-presenting cell by controlling the zinc ion concentration was reversible, and the present invention was completed. That is, the present invention is as follows. [1] A function-controlling agent for antigen-presenting cells, containing as an active ingredient a substance capable of controlling intracellular zinc ion concentration.
  • Zinc ion chelator is 2, 3-dimercapto-1 monopropanesulfonic acid (DMP S), N, N, ⁇ ', ⁇ , tetrakis (2-pyridylmethyl) ethylene diamine ( ⁇ ), ethylene diamine (EDTA) ) And ⁇ - (6-Methoxy-1-8-quinolyl) - ⁇ -toluenesulfonamide (TSQ).
  • DMP S 2, 3-dimercapto-1 monopropanesulfonic acid
  • N N, N, ⁇ ', ⁇ , tetrakis (2-pyridylmethyl) ethylene diamine ( ⁇ ), ethylene diamine (EDTA) )
  • TSQ ⁇ - (6-Methoxy-1-8-quinolyl) - ⁇ -toluenesulfonamide
  • Zinc ion-requiring protein is R af -1, PKC a, GE F— H l, H DAC, SQSTM1, ubiquitin monoprotein ligase E3, ubiquitin-coupled protein E2, metamouth thionein, phosphatase, Snai1, TRAF6, Paxi 1 1 in, Z yxin and J ade-1
  • the antigen-presenting cell is at least one selected from the group consisting of dendritic cells, macrophages, Langerhans cells, B cells, thymic epithelial cells, synovial cells, vascular endothelial cells, and keratinocytes, [1] To [9] The function control agent according to any one of [9].
  • a prophylactic / therapeutic agent for a disease involving an immune system comprising the function control agent according to any one of [1] to (: 1 1) above.
  • [1 3] Diseases that involve the immune system consist of autoimmune diseases, allergic diseases, rejection during organ transplantation, decreased immune response due to infection and tumor formation, and abnormal immune response when using vaccines
  • the prophylactic / therapeutic agent according to [12] above which is at least one selected from the group.
  • a prophylactic / therapeutic agent for diseases involving the immune system containing as an active ingredient a substance that can control intracellular zinc ion concentration.
  • the substance capable of controlling the zinc ion concentration is a zinc ion chelator, [14] The preventive / therapeutic agent as described.
  • the zinc ion chelator is 2,3-dimercapto-1- 1-propanesulfonic acid (DMP S), N, N, N, N, monotetrakis (2-pyridylmethyl) ethylenediamine (TPEN), ethylenediamine (EDTA) ) And N- (6-Methoxy-8-quinolyl) 1-p-toluenesulfonamide (T SQ), which is at least one selected from the group described in [17] above. medicine.
  • DMP S 2,3-dimercapto-1- 1-propanesulfonic acid
  • TPEN 2,3-dimercapto-1- 1-propanesulfonic acid
  • TPEN 2,3-dimercapto-1- 1-propanesulfonic acid
  • TPEN 2,3-dimercapto-1- 1-propanesulfonic acid
  • TPEN 2,3-dimercapto-1- 1-propanesulfonic acid
  • TPEN 2,3-dimercapto-1- 1-propanes
  • Zinc ion-requiring protein is Raf 1-1, PKCct, GEF— ⁇ 1, 'HDAC, SQSTM1, ubiquitin monoprotein ligase E3, ubiquitin-synthesizing protein E2, metamouthoneone, phosphatase, Snai1,
  • the prophylactic / therapeutic agent according to [19] above which is at least one selected from the group consisting of TRA F 6, Paxi 1 1 in, Z yxin and Jade-1.
  • the disease involving the immune system consists of autoimmune diseases, allergic diseases, rejection at the time of organ transplantation, decreased immune response due to infection and tumor formation, and abnormal immune response when using vaccines
  • the prophylactic / therapeutic agent according to any one of [14] to [22], which is at least one selected.
  • the antigen-presenting cell is at least one selected from the group consisting of dendritic cells, macrophages, Langerhans cells, B cells, thymic epithelial cells, synovial cells, vascular endothelial cells, and keratinocytes.
  • the prophylactic / therapeutic agent according to any of [23].
  • the zinc ion chelator is 2,3-dimercapto-1- 1-propanesulfonic acid (DMP S), N, N, ⁇ ', N'-tetrakis (2-pyridylmethyl) ethylenediamine ( ⁇ ), ethylenediamine (EDTA) and The method according to [29] above, which is at least one selected from the group consisting of ⁇ — (6-methoxy-8-quinolyl) 1 ⁇ -toluenesulfonamide (TSQ).
  • DMP S 2,3-dimercapto-1- 1-propanesulfonic acid
  • 2,3-dimercapto-1- 1-propanesulfonic acid
  • 2,3-dimercapto-1- 1-propanesulfonic acid
  • 2,3-dimercapto-1- 1-propanesulfonic acid
  • 2,3-dimercapto-1- 1-propanesulfonic acid
  • 2,3-dimercapto-1-
  • Zinc ion-requiring protein is Raf-1, PKCa, GEF-Hl, HDAC, SQSTM1, ubiquitin monoprotein ligase E3, ubiquitin synergistic protein E2, metamouth thionein, phosphatase, Snai1,
  • a method for screening a compound capable of controlling the function of antigen-presenting cells comprising measuring intracellular zinc ion concentration.
  • the antigen presenting cell is at least one selected from the group consisting of dendritic cells, macrophages, Langerhans cells, B cells, thymic epithelial cells, synovial cells, vascular endothelial cells, and keratinocytes, [3 6] The method described.
  • [4 1] Use of a substance capable of controlling the intracellular zinc ion concentration for the manufacture of a drug for the prevention and treatment of diseases involving the immune system.
  • FIG. 1 shows that when dendritic cells are treated with a zinc ion chelator, the expression of MH C class I, class I I, and CD 86 molecules on the cell surface is enhanced.
  • FIG. 2 shows that when dendritic cells are treated with zinc ions and zinc ionophores, LPS-stimulated dendritic cells enhance the expression of MH CII and CD86 molecules and suppress IL-12 production.
  • FIG. 1 shows that when dendritic cells are treated with a zinc ion chelator, the expression of MH C class I, class I I, and CD 86 molecules on the cell surface is enhanced.
  • Fig. 2 shows that when dendritic cells are treated with zinc ions and zinc ionophores, LPS-stimulated dendritic cells enhance the expression of MH CII and CD86 molecules and suppress IL-12 production.
  • FIG. 3 is a graph showing that dendritic cells in which m L i v 1 is expressed using a retroviral vector suppress the enhancement of MHC class I I molecule expression by L PS stimulation.
  • control of zinc ion concentration means in addition to the literal meaning of increasing or decreasing the concentration (amount) of zinc ion in the cell, and finally the zinc ion concentration increases or decreases in the cell. Induces a phenomenon similar to that caused by In such a case, it is not restricted by the concentration of zinc ions in the cell.
  • the cells to be controlled for zinc ion concentration include dendritic cells, macrophages, Lange / rehans cells, B cells, thymic epithelial cells and other antigen-presenting cells, as well as synovial cells, vascular endothelial cells, These cells are expected to acquire the ability to present antigens such as keratinocytes. Dendritic cells are preferred. In the present specification, the cells to be controlled are sometimes referred to as “antigen-presenting cells” for convenience. '
  • “Substances that can control intracellular zinc ion concentration” include “zinc ions”, “zinc ion chelators”, “zinc ionophores”, “substances that regulate the expression and Z or function of zinc ion-requiring proteins”, “ Examples include substances that regulate the expression and / or function of zinc ion transporters, etc. ⁇ Zinc ion J or zinc ionophore '' can directly increase the concentration of zinc ions in antigen-presenting cells by adding them.
  • the “zinc ion chelator” can directly reduce the zinc ion concentration in the antigen-presenting cell by adding it, thereby controlling the function of the antigen-presenting cell.
  • Substances that regulate the expression and ⁇ or function of zinc ion-requiring protein and “Substances that regulate the expression and / or function of zinc ion transporter” regulate the expression and ⁇ or function of zinc ion-requiring protein. Or by regulating the expression and ⁇ or function of the zinc ion transporter, it is possible to control the function of antigen-presenting cells by changing the effect of zinc ions on antigen-presenting cells. .
  • Substance that regulates the expression or function in a direction that promotes the expression or function of a substance that regulates the expression and function of a zinc ion-requiring protein or a substance that regulates the expression and / or function of a zinc ion transporter Then, the responsiveness to the zinc ion of the cell can be further enhanced, and the responsiveness to the zinc ion of the cell can be further suppressed as long as it is a substance that regulates the expression or function.
  • “control” of the zinc ion concentration means both positive and negative regulation.
  • Antigen-presenting cells consist of 1) foreign substances (foreign substances) derived from infection, etc., and 2) self Alternatively, endogenous proteins resulting from cell tumorigenesis, virus infection, etc. should be presented to cells with TCR (eg CD4 + T cells, CD8 + T cells, DNT cells, NKT cells, etc.). Therefore, we will protect against infection and remove cells that are undesirable for our by distinguishing between self and non-self.
  • TCR eg CD4 + T cells, CD8 + T cells, DNT cells, NKT cells, etc.
  • “Quality” here means response control and immune tolerance of T h 1 and T h 2 cells in CD 4+ T cells, and response of CTL with antigen-specific cytotoxic activity in CD 8 + T cells. It means control and immune tolerance, etc. “Amount J literally means strength of immune response. Also,“ controlling ”the function of an antigen-presenting cell means activation of the antigen-presenting cell itself or It means inactivation.
  • MHC class I and II molecules 1) expression control of MHC class I and II molecules, 2) expression control of costimulatory molecules such as CD 80, CD86, and CD 40, 3) IL-1, IL-6, IL-10, IL-1 2, IFN-a / j3, TNF_a, TGF—, etc., control of the expression of site force-in, 4) control of uptake of foreign or endogenous antigen, 5) control of migration ability of antigen-presenting cells, etc. Means to control the function of one or more items.
  • Zinc ions are introduced into antigen-presenting cells by zinc ionophores.
  • zinc ions are introduced into antigen-presenting cells in the form of a complex with a zinc ionophore.
  • Zinc ionophores include various compounds that are commonly used in the art, and preferably are commercially available. Examples include zinc pyrithione, heterocyclic amine, dithiol rubamate, and vitamins.
  • a “zinc ion chelator” is a substance that can remove zinc ions from cells by forming a complex with zinc ions in antigen-presenting cells.
  • DMP S 2, 3-dimercapto-1 Propanesulfonic acid
  • N, N, ⁇ ', N' Tetrakis (2-pyridylmethyl) Ethylenediamine ( ⁇ ), Ethylenediamine (EDTA) and ⁇ — (6-Methoxy 8-quinolyl) ⁇ -Toluenesulfonamide (T SQ) and the like. Both are commercially available.
  • a prophylactic / therapeutic drug for a disease involving the immune system of the present invention (hereinafter sometimes simply referred to as a prophylactic / therapeutic drug).
  • the amount of zinc ion chelator as a substance capable of controlling the zinc ion concentration is appropriately determined within a range in which the intracellular zinc ion concentration can be controlled.
  • One zinc ion chelator may be included, or two or more zinc ion chelators may be included. When two or more types are included, the blending amount is appropriately set according to the type.
  • “Substances that regulate the expression and Z or function of zinc ion-requiring protein” are not particularly limited as long as the expression and / or function of zinc ion-requiring protein can be regulated as a result. It may be regulation at the gene level or regulation at the protein level. Examples of regulation at the gene level include transcription regulation and gene expression regulation. Examples of regulation at the protein level include metabolic regulation, phosphorylation, dephosphorylation, sugar addition, lipid addition, coordinate bond with zinc, degradation, ubiquitination, acetylation, and the like. “Zinc ion-requiring protein” is a protein that requires zinc for the functional expression of antigen-presenting cells.
  • a protein having a zinc finger examples include proteins having fingers.
  • a zinc finger is a unique nucleic acid-binding motif that can have a higher-order structure only when an amino acid such as cystine-histidine is coordinated with zinc.
  • proteins having a zinc finger include epithelium.
  • a phenomenon called mesenchymal transition (such as body shaping in the early development of humans and other organisms, wound healing, cancer metastasis, etc.) Examples include transcription factors Snai1, PKCa, GEF—Hl, HDAC, SQSTM1, etc., which are master regulators of the phenomenon of releasing adhesion, acquiring mobility and moving to another).
  • the RI NG finger is a variant of the zinc finger, and is suggested to be involved in protein-protein interactions, and ubiquitin monoprotein ligase E 3 (ubiquitin np roteinligase) is a protein with a RI NG finger.
  • E 3 Active or ubiquitous It has been reported that it binds to conjugated nucleoprotein E 2 (ubiquitin—conjugating enzymes E 2) and TRAF 6 (TNF ⁇ signaling molecule).
  • the L IM domain consists of 60 amino acids in which the positions of 6 cysteines and 1 histidine are conserved. Although it has a structure similar to that of zinc fingers, its DNA-binding ability is unknown. It also functions as a domain that mediates binding to PKC.
  • proteins having a LIM domain include Pa'x i 11 in, Z yxin, etc. involved in cytoskeletal regulation.
  • the PHD zinc finger is a zinc finger uniform domain, a nuclear protein that is implicated in chromatin-mediated transcriptional regulation, and is predicted to have DNA-binding ability.
  • Examples of the protein having a PHD zinc finger include Jade-1 involved in histone acetylation.
  • the “substance that regulates the expression and function of zinc ion-requiring protein” may be a known compound or a new compound that will be developed in the future. Further, it may be a low molecular compound or a high molecular compound.
  • the low molecular weight compound is a compound having a molecular weight of less than about 3000, and includes, for example, organic compounds that can be normally used as pharmaceuticals and their derivatives and inorganic compounds. Derivatives, naturally-occurring compounds and derivatives thereof, small nucleic acid molecules such as promoters, various metals, and the like, desirably organic compounds that can be used as pharmaceuticals, derivatives thereof, and nucleic acid molecules.
  • the polymer compound is a compound having a molecular weight of about 3000 or more, and includes proteins, polynucleic acids, polysaccharides, and combinations thereof, and is preferably a protein.
  • These low-molecular compounds or high-molecular compounds are commercially available if they are known, or can be obtained through steps such as collection, production, and purification according to each report literature. These may be of natural origin, prepared by genetic engineering, or obtained by semisynthesis.
  • the expression of zinc auxotrophic protein S nai 1 is regulated by MAPK through TGF-] 3 or FGF (Peinado, H., Quintanilla,. & Cano, A.
  • Transforming growth factor beta-1 induces snail transcription factor in epithelial cell lines: mechanisms for epithelial mesenchymal transitions. J. Biol. Chem. 278, 21113-23 (2003)).
  • Snai 1 is activated by LIV 1 (described later), which is a zinc ion transporter (Yamashita, S., Miyagi, C., Fukada, ⁇ ⁇ , Kagara, ⁇ ., Che, ⁇ , -S. & Hirano, ⁇ .
  • Zinc transporter LIVI controls
  • Raf 1 For other zinc ion-requiring proteins, Raf 1, GEF— ⁇ ⁇ , PKC, phosphatase, and meta-mouthonein are also available.
  • SQS TM 1 and HD AC can be used as an example Joung I, Strominger JL, Shin J. Molecular cloning of a phosphotyrosine-independent ligand of the p561ck SH2 domain. Proc Natl Acad Sci USA, 93, 5991-5995 ( 1996) and Kawaguchi Y, Kovacs JJ, McLaurin A, Vance JM, Ito A, Yao TP.The deacetylase HDAC6 regulates aggresome formation and cell viability in response to misfolded protein stress.Cell 115, 727-738 (2003). Has been.
  • a substance that regulates the expression and / or function of zinc ion transporter means that the mechanism of action is not particularly limited as long as the expression and Z or function of zinc ion transporter can be regulated as a result. It may be at the level or at the protein level. Examples of regulation at the gene level include transcription regulation and gene expression regulation. Examples of regulation at the protein level include metabolic regulation, phosphorylation, dephosphorylation, sugar addition, lipid addition, coordinate bond with zinc, degradation, ubiquitination, acetylation, and the like.
  • Zero ion transporters include, for example, human ZIPs containing LIV family (BAB 70848, h Z ip 4, BI GM1 03, KI AA006 2, KI AA1 265, h L iv—1, A AH 088 53, hKE 4, XP— 20864 9, h ZIP l, h ZIP 2, h ZIP 3, BAA 9 2 100, BAC04504, etc., human CDFs (h Z nt _ 5, h Z nt— 7 HZnt-1, 1, hZnt-6, hZnt-3, hZnt-8, hZnt-4, hZnt-9, etc.).
  • LIV 1 was originally identified as an estrogen-controlled breast cancer protein, and recently the Zinc ion transporter subfamily (Z rt) called LZT (LIV 1 subfamily of ZIP zinc ion transporters).
  • Z rt Zinc ion transporter subfamily
  • ⁇ I rt The LZT proteins; the LIV-1 subfamily of zinc transporters. Biochim. Biophys. Acta 1611, 16-30 (2003)
  • function as a sub-ion ion transporter (Taylor, K.., Morgan, HE, Johnson, A., Hadley, Shi J. & Nicholson, RI
  • LIV-1 Structure-function analysis of LIV-1, tne breast cancer-associated protein that belongs to a new subfamily of zinc transporters. Biochem. J. 375, 51-9 (2003)).
  • Other examples include C DF (c a t i o n d i f f u s i o nf a c i 1 i tat o r) and the like.
  • the zinc ion transporter LIV 1 is regulated by STAT 3 (Yamashita, S., iyagi, C., Fukada, ⁇ . ', Kagara, N., Che, Y.-S. & Hirano , T.
  • Zinc transporter LIVI controls epithelial-mesenchymal transition in zebraf ish gastrula organizer. Nature 429, 298-302 (2004)).
  • the “substance that regulates the expression and function of zinc ion transporter” may be a known compound or a new compound to be developed in the future. Further, it may be a low molecular compound or a high molecular compound.
  • the low molecular weight compound is a compound having a molecular weight of less than about 300, for example, an organic compound that can be usually used as a pharmaceutical, a derivative thereof, and an inorganic compound.
  • the polymer compound is a compound having a molecular weight of about 300 or more, and includes proteins, polynucleic acids, polysaccharides, and combinations thereof, and is preferably a protein.
  • These low-molecular compounds or high-molecular compounds are commercially available if they are known, or can be obtained through steps such as collection, production, and purification according to each report literature. These may be of natural origin, prepared by genetic engineering, or obtained by semisynthesis.
  • LIV 1 a zinc ion transporter, is a zinc ion-requiring protein W
  • DNA encoding a protein having the amino acid sequence described in SEQ ID NO: 2, 4 or 6 (LIV 1 protein)-b) DNA comprising the sequence described in SEQ ID NO: 1, 3 or 5 (LIV 1 gene) c) DNA encoding a protein (protein similar to LIV 1) having an amino acid sequence in which one or more amino acid sequences are substituted, deleted, inserted and Z or added in the amino acid sequence described in SEQ ID NO: 2, 4 or 6
  • DN A consisting of the sequence described in SEQ ID NO: 1, 3 or 5
  • Double-stranded RNA having a sequence identical or similar to a part of DNA consisting of the sequence described in SEQ ID NO: 1, 3 or 5
  • (1) to (3) are substances that positively regulate the expression and / or function of Snai 1 which is a zinc ion-requiring protein (enhance expression and activate Z or function) (4 ) And (5) are substances that negatively regulate the expression and / or function of Snai 1 (reduce expression and / or suppress function).
  • the LIV 1 protein can be prepared by various methods well known to those skilled in the art. For example, it can be prepared by producing and purifying a protein in a transformant holding a vector having the DNA (LVI gene) having the nucleotide sequence set forth in SEQ ID NO: 1, 3 or 5 inserted therein.
  • the vector to be used can be appropriately selected depending on the translation system used for protein production.
  • LIV 1 is known to be expressed in hormonal tissues such as breast, prostate, pituitary gland, and brain (Taylor, KM & Nicholson, R. ⁇ .
  • Around anti-LIV 1 protein antibody LIV 1 protein can be purified from a cell extract expressing LIV 1 by preparing it by an intelligent method and preparing a affinity column with the antibody.
  • a protein similar to LIV 1 is a protein capable of regulating the activity of a zinc ion-requiring protein (for example, S nai 1), for example, one or more amino acids in the amino acid sequence set forth in SEQ ID NO: 2, 4 or 6 DNA that hybridizes under stringent conditions with a protein consisting of an amino acid sequence with substitution, deletion, insertion, and addition or deletion, or with a DNA consisting of a base sequence according to SEQ ID NO: 1, 3 or 5
  • S nai 1 zinc ion-requiring protein
  • a transformant is prepared using the obtained highly homologous DNA
  • a method can be used in which the transformant produces the desired protein.
  • a method for obtaining highly homologous DNA a part of the base sequence described in SEQ ID NO: 1, 3 or 5 is used as a probe, and stringent hybrids are obtained from cells of vertebrate animals other than human rabbits.
  • An example is a method of hybridizing under dithering conditions.
  • a hybrid solution containing 25% formamide, 50% formamide under more severe conditions 4 XS SC, 50 mM Hepes pH 7.0, 10 X Denhardt's solution, 20 8 11 and denatured salmon sperm 0
  • After prehybridization at 42 ° C add labeled probe and incubate at 42 ° C for high prehybridization.
  • the cleaning solution and temperature conditions are about ⁇ 1 XSSC, 0.1% SDS, 37 ° C '', and more severe conditions are ⁇ 0.5 XSSC, 0.1% SDS, 42 ° CJ, More severe conditions can be implemented at “0.2 XS SC, 0.1% SDS, 65 ° C”.
  • SSC SSC
  • SD S and temperature conditions A person skilled in the art can appropriately combine the above or other factors (for example, probe concentration, probe length, hybridization reaction time, etc.) that determine the stringency of the hybridization. It is possible to achieve the same stringency as
  • a polypeptide encoded by DNA isolated using such a hybridization technique usually has a high homology in amino acid sequence with L I V 1.
  • High compatibility means at least 40% or more, preferably 60% or more, more preferably 80% or more, more preferably 90% or more, still more preferably 95% or more, and particularly preferably 97% or more. This refers to the sequence homology (for example, 98 to 99%).
  • the identity of amino acid sequences can be determined by, for example, the algorithm B LAST (Proc. Natl. Ac ad. S ci. USA 8 7: 2264-2268, 1 990, Proc. Na tl. Ac a d. S c in USA 90: 587 3-58 77, 1 9 3 3).
  • B LAS TX Alt s c hul eta 1. J. Mo 1. B i o l. 2 1 5: 40 3— 4 10, 1 990).
  • BLAST and Gappe ED BLAST programs use the default parameters of each program. Specific methods of these analysis methods are publicly known, http: ZZwww.ncbi.n1m.nih.gov.).
  • Preparation of a protein similar to L I V 1 can also be performed by other known means.
  • DNA encodes the protein LIV 1, and if introduced into a cell, it is a substance that can indirectly adjust the activity of a zinc-requiring protein, such as Snai 1, through LIV 1 protein expression. .
  • the above DNA should be prepared from cDNA of cells expressing LIV 1 by a hybridization technique well known to those skilled in the art using a part of the sequence shown in SEQ ID NO: 1, 3 or 5 as a probe. Can do. It can also be obtained by performing RT-PCR from mRNA using a part of the sequence shown in SEQ ID NO: 1, 3 or 5 as a primer. Alternatively, it may be artificially synthesized using a commercially available DNA synthesizer.
  • DNA similar to the above DNA is also an example of “a substance that regulates the expression and function of a zinc ion-requiring protein”.
  • the similar DNA is a DNA that hybridizes under stringent conditions with the sequence shown in SEQ ID NO: 1, 3, or 5, and is a substance that regulates the expression and / or function of a zinc ion-requiring protein, for example,
  • a DNA encoding a protein capable of regulating the activity of Snai 1 can be mentioned. The method for preparing this DNA is as described above.
  • each DNA described above When using each DNA described above, it can be used by inserting it into an appropriate vector.
  • a product inserted into a vector is also one embodiment of the present invention.
  • the vector to be used can select an appropriate vector according to the purpose. Specifically, vectors derived from mammals (for example, pc DNA 3 (manufactured by Invitrogene) and pEG F- BOS (Nucleic Acids. Res., 1 8 (17), p.
  • insect cell-derived vectors eg “B ac-to- BAC baculovirus expression system” (Invitrogen), p B ac PAK8), plant-derived expression vectors 1 (for example, ⁇ 1, pMH2), a vector derived from an animal virus (for example, p HSV, pMV, pAd ex L cw), a vector derived from retrowinoles (for example, p ZIP neo), a vector derived from yeast (for example, “P ichia E xpression Kit J (manufactured by Invitrogen), pNV ll, SP—Q0 1), vectors derived from Bacillus subtilis (eg, p PL 608, p KTH 50), E.
  • Bacillus subtilis eg, p PL 608, p KTH 50
  • E Bacillus subtilis
  • coli vector M 1 3 Vector vectors, pUC vectors, pBR322, pBluescript ⁇ pCRScript) and the like.
  • an expression vector for example, calcium phosphate method (Virology, Vol. 52, p. 456, 1 973), DEAE dextran method, Cationic ribosome DOTAP (Roche. Diagnostics) , Electo mouth position method (Nuc 1 eic Acids Res., Vo l. 1 5, p. 1 3 1 1, 1 98 7), lipofection method (J. C 1 B ioch em.
  • Antisense oligonucleotides to DNA encoding L I V 1 are thought to prevent expression of the endogenous L I V 1 gene and negatively regulate Snai 1 activity.
  • Examples of such antisense oligonucleotides include antisense oligonucleotides having a DNA sequence consisting of the sequence set forth in SEQ ID NO: 1, 3 or 5 or an mRNA sequence generated from the DNA sequence as a target sequence.
  • the oligonucleotide described in SEQ ID NO: 7 or 8 can be mentioned.
  • it is included in the antisense oligonucleotide of the present invention as long as it hybridizes to any location such as DNA consisting of the sequence set forth in SEQ ID NO: 1, 3 or 5 and can effectively inhibit LIV 1 expression. It does not have to be completely complementary to DNA consisting of the sequence set forth in SEQ ID NO: 1, 3 or 5 or the corresponding mRNA.
  • the antisense oligonucleotide can be used by inserting it into an appropriate vector depending on the purpose.
  • retrovirus vector for the purpose of gene therapy, retrovirus vector, adenovirus vector, vaccinia virus vector, etc. It can be appropriately selected from viral vectors, non-viral vectors such as cationic ribosomes and ligand DNA complexes. It is also conceivable to administer a large volume of aqueous solution as naked plasmid DNA without using a carrier.
  • RNA i refers to a phenomenon in which when a double-stranded RNA (dsRNA) is introduced into a cell, the mRNA in the cell corresponding to the RNA sequence is specifically degraded and is not expressed as a protein.
  • dsRNA double-stranded RNA
  • the region that forms a double strand may form a double strand in all regions, or a part of the region (for example, both ends or one end) may be a double strand or the like.
  • the double-stranded RNA of the present invention may contain a region that is not double-stranded.
  • Oligo RNA used for RNA i is often 10 to: L 00 bp RNA, and usually 19 to 23 bp RNA.
  • RNAi method is Nature, Vo l. 39 1, p. 806, 1 9 98, Proc. N atl. Ac ad. S ci. USA Vo l. 95, p. 1 5502, 1 998, N ature, Vol. 395, p. 854, 1 998, Proc.
  • the “Zinc ion transporter” is other than LIV 1, for example, h Z nt— 1, h Z nt— 2, h Z nt— 5, h Z nt— 6, h Z nt— 7, h ZIP l, h ZIP 2, h ZIP 3, BAA 9 2 1 00, BAC 04504, h L iv— 1, hKE 4, KI AA 1 265, KI AA 0062, h Z ip 4
  • a substance that modulates Z or function can be prepared. -Information on various amino acid sequences and nucleotide sequences can be obtained from various databases that can be browsed by NCB I.
  • the agent for controlling the function of antigen-presenting cells and the prevention of diseases involving the immune system includes the above-described series of active ingredients (zinc ion, zinc ionophore, zinc ion killer, expression of zinc ion-requiring protein) And substances that regulate Z or function, substances that regulate the expression and function of zinc ion transporter), and optionally include pharmaceutically acceptable excipients and additives.
  • Pharmaceutically unacceptable excipients and additives include carriers, binders, fragrances, buffers, thickeners, colorants, stabilizers, emulsifiers, dispersants, suspending agents, preservatives, etc. Can be mentioned.
  • different types of “substances that control zinc ion concentration” can be used in combination as active ingredients.
  • Examples of pharmaceutically acceptable carriers include magnesium carbonate, magnesium stearate, tanolec, sugar, ratatoose, pectin, dextrin, starch, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose, and low melting point.
  • Examples include wax and cocoa butter.
  • the tablets can be made into tablets with ordinary coatings as necessary, for example, sugar-coated tablets, enteric-coated tablets, film-coated tablets, double-layer tablets, and multilayer tablets.
  • Powders are formulated with a pharmaceutically acceptable powder base. Examples of bases include talc, lactose, and starch. Drops are aqueous or non-aqueous It can be formulated together with a base and one or more pharmaceutically acceptable diffusing agents, suspending agents, solubilizing agents and the like.
  • Capsules can be produced by filling a compound as an active ingredient together with a pharmaceutically acceptable carrier. The compounds can be mixed with pharmaceutically acceptable excipients or filled into force capsules without excipients. A cachet agent can be produced by a similar method.
  • the present invention When the present invention is prepared as a suppository, it is usually used together with a base such as vegetable oil (castor oil, olive oil, peanut oil, etc.), mineral oil (petrol, white petrolatum, etc.), waxes, 'partially synthesized or fully synthetic glycerin fatty acid ester. Formulated according to the technique used.
  • a base such as vegetable oil (castor oil, olive oil, peanut oil, etc.), mineral oil (petrol, white petrolatum, etc.), waxes, 'partially synthesized or fully synthetic glycerin fatty acid ester.
  • injection solutions include solutions, suspensions, and emulsions.
  • an aqueous solution, a water-propylene glycol solution, etc. are mentioned.
  • the solution can also be produced in the form of a solution of polyethylene and / or propylene dalicol, which may contain water.
  • a solution suitable for oral administration can be produced by adding a compound as an active ingredient to water and adding a colorant, a fragrance, a stabilizer, a sweetener, a solubilizer, a thickener and the like as necessary.
  • a solution suitable for oral administration can also be produced by adding the compound together with a dispersant to water to make it viscous.
  • the thickener include pharmaceutically acceptable natural or synthetic gum, resin, methylcellulose, sodium carboxymethylcellulose, or a known suspending agent.
  • topical administration agent examples include the above-mentioned liquids, creams, aerosols, sprays, powders, lotions, ointments and the like.
  • the above-mentioned topical preparation can be produced by mixing a compound as an active ingredient with a pharmaceutically acceptable diluent and carrier.
  • Ointments and creams are formulated, for example, by adding a thickener and a Z or gelling agent to an aqueous or oily base.
  • the base include water, liquid paraffin, and vegetable oil.
  • thickening agent examples include soft paraffin, anolenomium stearate, cetostellenoreanolol, propylene glycolate, polyethylene glycol, lanolin, hydrogenated lanolin, and beeswax.
  • methyl hydroxybenzoate, propyl hydroxybenzoate, Preservatives such as black mouth cresol and benzalkonium chloride and bacterial growth inhibitors can be added.
  • Lotions can contain one or more pharmaceutically acceptable stabilizers, suspending agents, emulsifiers, diffusing agents, thickeners, colorants, flavors, etc., in an aqueous or oily base. .
  • the antigen-presenting cell function-controlling agent of the present invention and the prophylactic / therapeutic agent for diseases related to the immune system are administered orally or parenterally.
  • a dosage form When administered orally, it can be administered in a dosage form commonly used in the art. When administered parenterally, it can be administered in a dosage form such as a topical administration agent (transdermal agent, etc.), a rectal agent, an injection, or a nasal agent.
  • a topical administration agent transdermal agent, etc.
  • a rectal agent an injection, or a nasal agent.
  • oral preparation or rectal preparation examples include capsules, tablets, pills, powders, drops, cachets, suppositories, and liquids.
  • injections examples include sterile solutions or suspensions.
  • topical agents include creams, ointments, lotions, transdermal agents (ordinary patches, matrix agents) and the like.
  • the dose and the number of doses vary depending on the type of substance that can control the zinc ion concentration to be used, the patient's symptoms, age, weight, dosage form, etc., and can be appropriately set.
  • Zinc ions In mammals such as humans, horses, horses, dogs, mice, rats, etc., the function of antigen-presenting cells, specifically the expression of molecules related to antigen presentation, the expression of antigen-presenting cells by LPS, etc. It is possible to control activation, site force-in production, antigen uptake, migration ability, etc., and therefore prevent diseases related to the function of antigen-presenting cells, specifically diseases related to the immune system It can be treated.
  • the present invention includes SIRS (systemic inflammatory response syndrome), systemic lupus erythematosus, mixed connective tissue disease, rheumatoid arthritis, siedalene syndrome, rheumatic fever, Good Pastier I syndrome, Graves' disease, Hashimoto's disease, Addison's disease, autoimmune hemolytic anemia, idiopathic platelet-poor purpura, myasthenia gravis, ulcerative colitis, Crohn's disease, interchangeable ophthalmitis, multiple sclerosis To treat psoriasis, anaphylactic shock, allergic rhinitis, allergic conjunctivitis, bronchial asthma, measles, atopic dermatitis, etc.
  • SIRS systemic inflammatory response syndrome
  • systemic lupus erythematosus mixed connective tissue disease
  • rheumatoid arthritis siedalene syndrome
  • rheumatic fever Good Pastier I syndrome
  • Graves' disease Ha
  • Cancers include breast cancer, prostate cancer, knee cancer, stomach cancer, lung cancer, colon cancer (colon cancer, rectal cancer, anal cancer), esophageal cancer, duodenal cancer, head and neck cancer (tongue cancer, pharyngeal cancer), Brain tumor, schwannoma, non-small cell lung cancer, small cell lung cancer, liver cancer, kidney cancer, bile duct cancer, uterine cancer (uterine body cancer, cervical cancer), ovarian cancer, bladder cancer ', skin cancer, hemangioma, Malignant lymphoma, Malignant melanoma, Thyroid cancer, Bone tumor, Hemangioma, Angiofibroma, Retinal sarcoma, Penile cancer, Pediatric solid cancer, Force positive sarcoma, Capodisarcoma due to AIDS, Maxillary sinus tumor, Fibrous histiosphere Tumor, leiomyosarcoma, rhabdomyo
  • the agent for preventing and treating diseases associated with the function-controlling agent of the antigen-presenting cell and the immune system of the present invention suffers from various diseases whose therapeutic effect is recognized by the introduction of zinc ions into the antigen-presenting cell. Patients can also be the subject of administration.
  • antigen-presenting cells can be measured by measuring the intracellular zinc ion concentration. It is possible to screen for compounds that can control the functions of these. For example, the screening method is implemented by the following steps.
  • the antigen-presenting cells those described above are used. Dendritic cells are preferred. Appropriately divide into two groups and treat one with the test compound. The other is not treated and used as a control cell. Furthermore, compounds known to affect the zinc ion concentration, such as zinc ion chelator TPEN, may be used as a positive control compound.
  • the test compound may be a known compound or a new compound to be developed in the future. Further, it may be a low molecular compound or a high molecular compound.
  • the low molecular weight compound is a compound having a molecular weight of less than 300, for example, an organic compound that can be normally used as a pharmaceutical, a derivative thereof, and an inorganic compound.
  • the high molecular compound is a compound having a molecular weight of about 300 or more, and includes proteins, polynucleic acids, polysaccharides, and combinations thereof, and is preferably a protein.
  • These low-molecular compounds or high-molecular compounds can be obtained commercially if they are known, or can be obtained through steps such as collection, production, and purification according to each report literature. These may be of natural origin, may be prepared by genetic engineering, or may be obtained by semisynthesis or the like.
  • the treatment time of the cells with the test compound is appropriately set depending on the type of cells used and the test compound.
  • a positive control compound such as TPEN
  • the concentration of the test compound for treating the cells is also set appropriately depending on the type of cells used, the type of test compound, and the treatment time.
  • the measurement of intracellular zinc ion concentration can be carried out by using a method usually carried out in this field or according to it. For example, it can be measured directly by an atomic absorption method (frame method) or by a fluorescence spectrophotometer using a specific probe (for example, a fluorescent reagent).
  • Bone marrow cells were obtained from C 5 7 B LZ6 mice (obtained from 3 Claires), 10% FC S (EQU I TECH-B IOInc., # S FB—30— 1 388), granulocytes, macrophage colonies Cells were cultured in RPMI medium (SI GMA, R 87 58) with stimulating factor (GM—CSF) for 6 days at 37 ° C in a 5% CO 2 environment, and bone marrow-derived dendritic cells (BMDC) were cultured. Differentiation was induced. To this BMDC, TPEN (SI GM A, P 44 1 3) was added to the medium at a concentration of 0 to 2 M for 6 to 12 hours.
  • BMDC bone marrow-derived dendritic cells
  • LPS lipopolysaccharide
  • BMDC prepared in the same manner as in Example 1 was added with zincthiophore pyrithione (5 / M; Mollecularprobes, ⁇ —24 1 93) and Zn 2+ (Z n S0 4 ) in the final concentration of 5 In addition, it was stimulated with LPS (10 OngZmL '). Six hours later, MHC class II and CD86 molecules expressed on the cell surface were evaluated by FACS analysis. The result is shown in figure 2. The numbers in Figure 2 indicate the percentage of cells that highly express each molecule. At the same time, IL-12 production of BM DC was immobilized with 4% paraformaldehyde, followed by anti-IL-12 p40 rod (BD.
  • BD anti-IL-12 p40 rod
  • Bioscience P harmingen # 5 54 4 7 9) and isotype control antibody. (BD. Bioscience P harmingen, # 5 5 3 9 2 5) was used for detection by intracellular staining. The numbers in the figure show the percentage of cells producing IL-12.
  • MHC I I and CD 86 molecules on the cell surface and the production of IL-12 cytokines following activation of BMDC by LPS stimulation were not observed in the presence of zinc ionophore.
  • the mL i V 1 gene was cleaved and collected by I, and the University of Tokyo Medical Research Institute Produced by Prof. Toshio Kitamura and incorporated into the provided p MX—I RE S—GF P retrovirus vector.
  • This pMX—IRE S-GF P-mL iv 1 vector was introduced into Phoenix cells provided by Dr. Garry P. No 1 an at Stanford University, USA using lipofectamine (Invitrogen) to transfer the virus. Expressed.
  • GM-CSF GM-CSF producing cell line (GM-CSF / CHO) was cultured, and the culture supernatant was treated with 0.3% FCS (10%) and prot am inesu 1 fate (SI GMA; 2 ⁇ g / ml) were added to obtain a virus infection solution.
  • C 5 7 B LZ6 mouse (S-crea) BMDC prepared from bone marrow was infected with BM DC by replacing the medium with virus infection solution in Day 2 and Day 4. The medium was replaced with virus-free medium with Day 6, and further stimulated with LPS (SI GMA; 100 ng / mL).
  • Cells were collected 6 hours after stimulation, and the expression level of MHC class II molecules on the cell surface was examined by FACS.
  • a cell group not stimulated with LPS was treated as a control, and a GFP positive cell was treated as a cell in which mL i V 1 was forcibly expressed.
  • Sequence number 8 Antisense sequence
  • antigen-presenting cells molecules involved in antigen presentation (eg, MHC class I, class) by controlling the zinc ion concentration or regulating the expression and function of zinc ion-requiring proteins and zinc ion transporters II, CD 86 molecule, etc.) can be enhanced. Furthermore, introduction of zinc ions into antigen-presenting cells with a drug such as zinc ionophore can suppress activation of antigen-presenting cells and production of cyto force in by lipopolysaccharide (LPS).
  • LPS lipopolysaccharide

Abstract

Disclosed is an agent for controlling the function of an antigen-presenting cell. An agent for controlling the function of an antigen-presenting cell comprising, as an active ingredient, a substance capable of controlling the concentration of zinc ions in a cell, specifically a zinc ion, a zinc-ion chelator, a substance capable of controlling the expression and/or function of a zinc ion-requiring protein or a substance capable of controlling the expression and/or function of a zinc ion transporter.

Description

明細書 '- 抗原提示細胞の機能制御剤  Description '-Function control agent of antigen-presenting cell
技術分野  Technical field
本発明は抗原提示細胞の機能を制御し得る薬剤に関する。 より具体的には抗原 提示細胞の活性化を介した免疫応答反応を制御し得る薬剤に関する。  The present invention relates to a drug capable of controlling the function of antigen-presenting cells. More specifically, the present invention relates to a drug capable of controlling an immune response reaction through activation of antigen-presenting cells.
背景技術  Background art
人間の健康'を維持するシステムの一つに免疫系がある。 免疫応答の制御は感染 防御を目的としたワクチン開発、 自己免疫疾患の治療、 ガン等の克服、 臓器移植 の際の拒絶反応の抑制の点からも極めて重要である。 これまで、 この免疫応答の 制御を目的に様々な免疫賦活化剤、 例えば B C G等の菌体成分が使用されたり、 また菌体成分由来の免疫抑制剤や合成ステロイド剤が人体に投与されたりしてき た。 しかしながら、 これらの菌体成分や合成ステロイ ド剤は様々な副作用が存在 し、 人体にとって有害な一面を潜在的に持っている。 また長期使用によりその効 果が減弱する等、 薬剤として使用する際には、 これまでにいくつかの問題が指摘 されていた。  One system that maintains human health is the immune system. Control of the immune response is also extremely important from the viewpoint of vaccine development for the purpose of infection prevention, treatment of autoimmune diseases, overcoming cancer, etc., and suppression of rejection during organ transplantation. Until now, various immunostimulators such as BCG and other bacterial components have been used to control this immune response, and immunosuppressants and synthetic steroids derived from bacterial components have been administered to the human body. It was. However, these bacterial cell components and synthetic steroids have various side effects and potentially have a harmful aspect to the human body. In addition, some problems have been pointed out so far when it is used as a medicine, such as its effectiveness diminishing with long-term use.
樹状細胞等の抗原提示細胞は免疫応答の強さと質の制御の中心的存在であり、 自己免疫疾患、アレルギー性疾患、癌の免疫療法等への利用がすすめられている。 又、 抗原提示細胞の増殖や活性化を制御する培養法等の研究がすすめられている 力 依然、 効率よく且つ特異的に抗原提示細胞の機能を制御する方法が求められ ているのが現状である。  Antigen-presenting cells such as dendritic cells are central to the control of the strength and quality of the immune response, and are being used for autoimmune diseases, allergic diseases, cancer immunotherapy, and the like. In addition, research on culture methods that control the growth and activation of antigen-presenting cells is underway. Currently, there is still a need for a method that efficiently and specifically controls the function of antigen-presenting cells. is there.
従来技術 Iこつレヽて fま Guermonprez P, Val ladeau J, Zitvogel L, Thery C, Amigorena S. Antigen presentation and T cel l stimulation by denaritic cel ls. Annu Rev. Immunol. 20, 621-627 (2002)、 Banchereau J, Briere F, Caux C, Davoust J, Lebecque S, Liu YJ, Pulendran B, Palucka K. Immunobiology of dendritic cel ls. Annu Rev. Immunol. 18, 761-781 (2000)、 Steinman RM, ellman I. Prior art I Gutmonprez P, Val ladeau J, Zitvogel L, Thery C, Amigorena S. Antigen presentation and T cel l stimulation by denaritic cel ls. Annu Rev. Immunol. 20, 621-627 (2002), Banchereau J, Briere F, Caux C, Davoust J, Lebecque S, Liu YJ, Pulendran B, Palucka K. Immunobiology of dendritic cel ls.Annu Rev. Immunol. 18, 761-781 (2000), Steinman RM, ellman I.
Immunotherapy : bewitched, bothered, and bewi ldered no more. Science 305, 197-200 (2004)に詳述されている。 発明の開示 Immunotherapy: bewitched, bothered, and bewildered no more. Science 305, 197-200 (2004). Disclosure of the invention
本発明は、 新規な抗原提示細胞の機能制御剤の提供を目的とし、 具体的には抗 原提示細胞の活性化を介した免疫応答を制御 (ここで言う制御とは、 目的に応じ て増強と抑制を含む) し得る薬剤の提供を目的とする。 さらに本発明は、 当該薬 剤を自己免疫疾患の治療、 アレルギー性疾患の治療、 臓器移植の際の拒絶反応の 抑制等の各種疾患への適用、 さらに癌に対する免疫応答の増強法の開発や、 効率 的なワクチンの開発を目的とする。  An object of the present invention is to provide a novel function-controlling agent for antigen-presenting cells. Specifically, the present invention controls immune responses via activation of antigen-presenting cells. The purpose is to provide a drug capable of Furthermore, the present invention applies the drug to various diseases such as treatment of autoimmune diseases, treatment of allergic diseases, suppression of rejection during organ transplantation, and further development of a method for enhancing immune response against cancer, The purpose is to develop an efficient vaccine.
本発明者らは、 上記課題に鑑み、 鋭意検討を行った結果、 亜鉛イオンレベルを 制御することにより樹状細胞等の抗原提示細胞の機能を制御できることを見出し た。 さらに亜鉛イオン濃度の制御による抗原提示細胞の機能制御が可逆的である ことを確認して本発明を完成するに至った。すなわち本発明は以下の通りである。 〔1〕 細胞内の亜鉛イオン濃度を制御し得る物質を有効成分として含有する、 抗 原提示細胞の機能制御剤。  As a result of intensive studies in view of the above problems, the present inventors have found that the function of antigen-presenting cells such as dendritic cells can be controlled by controlling the zinc ion level. Furthermore, it was confirmed that the function control of the antigen-presenting cell by controlling the zinc ion concentration was reversible, and the present invention was completed. That is, the present invention is as follows. [1] A function-controlling agent for antigen-presenting cells, containing as an active ingredient a substance capable of controlling intracellular zinc ion concentration.
〔2〕 亜鉛イオン濃度を制御し得る物質が亜鉛イオンである、 上記 〔1〕 記載の 機能制御剤。  [2] The function control agent according to [1] above, wherein the substance capable of controlling the zinc ion concentration is zinc ion.
〔3〕 亜鉛イオンが亜鉛ィオノフォアによって細胞内に導入される、 上記 〔2〕 記載の機能制御剤。  [3] The function control agent according to the above [2], wherein the zinc ion is introduced into the cell by a zinc ionophore.
〔4〕亜鉛イオン濃度を制御し得る物質が亜鉛イオンキレ一ターである、上記〔 1〕 記載の機能制御剤。  [4] The function control agent according to the above [1], wherein the substance capable of controlling the zinc ion concentration is a zinc ion killer.
〔5〕 亜鉛イオンキレーターが 2, 3—ジメルカプト— 1一プロパンスルホン酸 (DMP S)、 N, N, Ν', Ν, ーテ トラキス (2—ピリジルメチル) エチレン ジァミン (ΤΡΕΝ)、 エチレンジァミン (EDTA) 及び Ν— (6—メ トキシ一 8—キノ リル) 一 ρ— トルエンスルホンァミ ド (T S Q) からなる群より選択さ れる少なくとも 1種である、 上記 〔4〕 記載の機能制御剤。  [5] Zinc ion chelator is 2, 3-dimercapto-1 monopropanesulfonic acid (DMP S), N, N, Ν ', Ν, tetrakis (2-pyridylmethyl) ethylene diamine (ΤΡΕΝ), ethylene diamine (EDTA) ) And Ν- (6-Methoxy-1-8-quinolyl) -ρ-toluenesulfonamide (TSQ). The function control agent according to [4] above, which is at least one selected from the group consisting of
〔6〕 ffi鉛イオン濃度を制御し得る物質が、 亜鉛イオン要求性タンパク質の発現 及び 又は機能を調節する物質である、 上記 〔1〕 記載の機能制御剤。  [6] The function control agent according to [1] above, wherein the substance capable of controlling the ffi lead ion concentration is a substance that regulates the expression and / or function of a zinc ion-requiring protein.
〔7〕 亜鉛イオン要求性タンパク質が R a f — 1、 PKC a、 GE F— H l、 H DAC、 SQSTM1、 ュビキチン一タンパク質リガーゼ E 3、 ュビキチン共役 タンパク質 E 2、 メタ口チォネイン、 フォスファターゼ、 S n a i 1、 TRAF 6、 P a x i 1 1 i n、 Z y x i n及び J a d e— 1からなる群より選択される 少なくとも 1種である、 上記 〔6〕 記載の機能制御剤。 [7] Zinc ion-requiring protein is R af -1, PKC a, GE F— H l, H DAC, SQSTM1, ubiquitin monoprotein ligase E3, ubiquitin-coupled protein E2, metamouth thionein, phosphatase, Snai1, TRAF6, Paxi 1 1 in, Z yxin and J ade-1 The function control agent according to [6] above, which is at least one kind.
〔8〕 亜鉛イオン濃度を制御し得る物質が、 亜鉛イオントランスポーターの発現 及び Z又は機能を調節する物質である、 上記 〔1〕 記載の機能制御剤。  [8] The function control agent according to [1] above, wherein the substance capable of controlling the zinc ion concentration is a substance that regulates expression and Z or function of a zinc ion transporter.
〔9〕 亜鉛イオントランスポーターが L I Vファミリ一を含むヒ ト Z I P類、 ヒ ト CDF類からなる群より選択される少なくとも 1種である、 上記 〔8〕 記載の 機能制御剤。  [9] The function control agent according to [8] above, wherein the zinc ion transporter is at least one selected from the group consisting of human Z IPs and human CDFs including the L I V family.
〔10〕 抗原提示細胞が樹状細胞、 マクロファージ、 ランゲルハンス細胞、 B細 胞、 胸腺上皮細胞、 滑膜細胞、 血管内皮細胞及びケラチノサイ トからなる群より 選ばれる少なくとも 1種である、 上記 〔1〕 〜 〔9〕 のいずれかに記載の機能制 御剤。  [10] The antigen-presenting cell is at least one selected from the group consisting of dendritic cells, macrophages, Langerhans cells, B cells, thymic epithelial cells, synovial cells, vascular endothelial cells, and keratinocytes, [1] To [9] The function control agent according to any one of [9].
〔1 1〕 抗原提示細胞が樹状細胞である、 上記 〔10〕 記載の機能制御剤。  [11] The function control agent according to [10] above, wherein the antigen-presenting cell is a dendritic cell.
〔1 2〕 上記 〔1〕 〜 (: 1 1〕 のいずれかに記載の機能制御剤を含む、 免疫シス テムが関与する疾患の予防 ·治療薬。  [1 2] A prophylactic / therapeutic agent for a disease involving an immune system, comprising the function control agent according to any one of [1] to (: 1 1) above.
〔1 3〕.免疫システムが関与する疾患が、 自己免疫疾患、 アレルギー性疾患、 臓 器移植の際の拒絶反応、 感染や腫瘍形成による免疫応答の低下及びワクチン使用 時の異常な免疫応答からなる群より選ばれる少なくとも 1種である、上記〔1 2〕 記載の予防 ·治療薬。  [1 3] Diseases that involve the immune system consist of autoimmune diseases, allergic diseases, rejection during organ transplantation, decreased immune response due to infection and tumor formation, and abnormal immune response when using vaccines The prophylactic / therapeutic agent according to [12] above, which is at least one selected from the group.
〔14] 細胞内の亜鉛イオン濃度を制御し得る物質を有効成分として含有する、 免疫システムが関与する疾患の予防 ·治療薬。  [14] A prophylactic / therapeutic agent for diseases involving the immune system, containing as an active ingredient a substance that can control intracellular zinc ion concentration.
〔1 5〕 亜鉛イオン濃度を制御し得る物質が亜鉛イオンである、 上記 〔1 4〕 記 載の予防 ·治療薬。  [15] The prophylactic / therapeutic agent according to [14] above, wherein the substance capable of controlling the zinc ion concentration is zinc ion.
〔1 6〕亜鉛イオンが亜鉛ィオノフォアによって細胞内に導入されるものである、 上記 〔1 5〕 記載の予防 ·治療薬。  [16] The prophylactic / therapeutic agent according to [15] above, wherein the zinc ion is introduced into the cell by a zinc ionophore.
〔1 7〕 亜鉛イオン濃度を制御し得る物質が亜鉛イオンキレーターである、 上記 〔14〕 記載の予防 ·治療薬。 [17] The substance capable of controlling the zinc ion concentration is a zinc ion chelator, [14] The preventive / therapeutic agent as described.
〔1 8〕 亜鉛イオンキレーターが 2, 3—ジメルカプト一 1—プロパンスルホン 酸 (DMP S)、 N, N, N,, N, 一テトラキス (2—ピリジルメチル) ェチレ ンジァミン (TPEN)、 エチレンジァミン (EDTA) 及び N— (6—メ トキシ —8—キノ リル) 一 p—トルエンスルホンアミ ド (T SQ) からなる群より選択 される少なく とも 1種である、 上記 〔1 7〕 記載の予防 ·治療薬。  [1 8] The zinc ion chelator is 2,3-dimercapto-1- 1-propanesulfonic acid (DMP S), N, N, N, N, monotetrakis (2-pyridylmethyl) ethylenediamine (TPEN), ethylenediamine (EDTA) ) And N- (6-Methoxy-8-quinolyl) 1-p-toluenesulfonamide (T SQ), which is at least one selected from the group described in [17] above. medicine.
〔1 9〕 亜鉛'イオン濃度を制御し得る物質が、 亜鉛イオン要求性タンパク質の発 現及び 又は機能を調節する物質である、 上記 〔 14〕 記載の予防 ·治療薬。 [19] The prophylactic / therapeutic agent according to [14] above, wherein the substance capable of controlling the zinc 'ion concentration is a substance that regulates the expression and / or function of a zinc ion-requiring protein.
〔20〕 亜鉛イオン要求性タンパク質が R a f 一 1、 PKCct、 GEF— Η 1、' HDAC、 SQSTM1、 ュビキチン一タンパク質リガーゼ E 3、 ュビキチン共 役タンパク質 E 2、 メタ口チォネイン、 フォスファターゼ、 S n a i 1、 TRA F 6、 P a x i 1 1 i n、 Z y x i n及び J a d e— 1からなる群より選択され る少なくとも 1種である、 上記 〔1 9〕 記載の予防 ·治療薬。 [20] Zinc ion-requiring protein is Raf 1-1, PKCct, GEF— Η1, 'HDAC, SQSTM1, ubiquitin monoprotein ligase E3, ubiquitin-synthesizing protein E2, metamouthoneone, phosphatase, Snai1, The prophylactic / therapeutic agent according to [19] above, which is at least one selected from the group consisting of TRA F 6, Paxi 1 1 in, Z yxin and Jade-1.
〔2 1〕 亜鈴イオン濃度を制御し得る物質が、 亜鉛イオントランスポーターの発 現及び/又は機能を調節する物質である、 上記 〔14〕 記載の予防 ·治療薬。 [2 1] The prophylactic / therapeutic agent according to [14] above, wherein the substance capable of controlling the dumbbell ion concentration is a substance that regulates the expression and / or function of a zinc ion transporter.
〔22〕 亜鉛イオントランスポーターが L I Vフアミリーを含むヒ ト Z I P類、 ヒ ト CD F類からなる群より選択される少なくとも 1種である、 上記 〔2 1〕 記 載の予防 ·治療薬。 [22] The prophylactic / therapeutic agent according to the above [2 1], wherein the zinc ion transporter is at least one selected from the group consisting of human Z IPs and human CD Fs containing LV family.
〔23〕 免疫システムが関与する疾患が、 自己免疫疾患、 アレルギー性疾患、 臓 器移植の際の拒絶反応、 感染や腫瘍形成による免疫応答の低下及びワクチン使用 時の異常な免疫応答からなる群より選ばれる少なく とも 1種である、上記〔14〕 〜 〔22〕 のいずれかに記載の予防 ·治療薬。  [23] The disease involving the immune system consists of autoimmune diseases, allergic diseases, rejection at the time of organ transplantation, decreased immune response due to infection and tumor formation, and abnormal immune response when using vaccines The prophylactic / therapeutic agent according to any one of [14] to [22], which is at least one selected.
〔24〕 抗原提示細胞が樹状細胞、 マクロファージ、 ランゲルハンス細胞、 B細 胞、 胸腺上皮細胞、 滑膜細胞、 血管内皮細胞及びケラチノサイ 卜からなる群より 選ばれる少なく とも 1種である、 上記 〔14〕 〜 〔23〕 のいずれかに記載の予 防 ·治療薬。  [24] The antigen-presenting cell is at least one selected from the group consisting of dendritic cells, macrophages, Langerhans cells, B cells, thymic epithelial cells, synovial cells, vascular endothelial cells, and keratinocytes. ] The prophylactic / therapeutic agent according to any of [23].
〔25〕 抗原提示細胞が樹状細胞である、 上記 〔24〕 記載の予防 ·治療薬。 〔26〕 インビトロで、 細胞内の亜鉛イオン濃度を制御することを含む、 抗原提 示細胞の機能を制御する方法。 [25] The prophylactic / therapeutic agent according to [24] above, wherein the antigen-presenting cell is a dendritic cell. [26] A method for controlling the function of an antigen-presenting cell, comprising controlling the intracellular zinc ion concentration in vitro.
〔27〕 亜鉛イオン濃度の制御が亜鉛イオンの細胞内への導入によって行われる ものである、 上記 〔26〕 記載の方法。  [27] The method according to [26] above, wherein the zinc ion concentration is controlled by introducing zinc ions into cells.
〔28〕 亜鋭イオンの細胞内への導入が亜鉛ィオノフォアによって行われるもの である、 上記 〔27〕.記載の方法。  [28] The method according to [27] above, wherein the subacute ion is introduced into the cell by a zinc ionophore.
〔29〕 亜鉛'イオン濃度の制御が亜鉛イオンキレーターによって行われるもので ある、 上記 〔26〕 記載の方法。  [29] The method described in [26] above, wherein the zinc 'ion concentration is controlled by a zinc ion chelator.
〔30〕 亜鉛イオンキレーターが 2, 3—ジメルカプト一 1—プロパンスルホン 酸 (DMP S)、 N, N, Ν', N' ーテトラキス (2—ピリジルメチル) ェチレ ンジァミン (ΤΡΕΝ)、 エチレンジァミン (EDTA) 及び Ν— (6—メ トキシ —8—キノリル) 一 ρ—トルエンスルホンアミ ド (T SQ) からなる群より選択 される少なくとも 1種である、 上記 〔29〕 記載の方法。  [30] The zinc ion chelator is 2,3-dimercapto-1- 1-propanesulfonic acid (DMP S), N, N, Ν ', N'-tetrakis (2-pyridylmethyl) ethylenediamine (ΤΡΕΝ), ethylenediamine (EDTA) and The method according to [29] above, which is at least one selected from the group consisting of Ν— (6-methoxy-8-quinolyl) 1 ρ-toluenesulfonamide (TSQ).
〔3 1〕 亜鉛イオン濃度の制御が亜鉛イオン要求性タンパク質の発現及びノ又は 機能を調節することによって行われるものである、 上記 〔26〕 記載の方法。 [31] The method according to [26] above, wherein the zinc ion concentration is controlled by regulating the expression and function of the zinc ion-requiring protein.
〔32〕 亜鉛イオン要求性タンパク質が R a f — 1、 PKC a、 GEF— H l、 HDAC、 SQSTM1、 ュビキチン一タンパク質リガーゼ E 3、 ュビキチン共 役タンパク質 E 2、 メタ口チォネイン、 フォスファターゼ、 S n a i 1、 TRA F 6, P a x i l 1 i n, Z y x i n及び J a d e - 1からなる群より選択され る少なくとも 1種である、 上記 〔3 1〕 記載の方法。 [32] Zinc ion-requiring protein is Raf-1, PKCa, GEF-Hl, HDAC, SQSTM1, ubiquitin monoprotein ligase E3, ubiquitin synergistic protein E2, metamouth thionein, phosphatase, Snai1, The method according to [3 1] above, which is at least one selected from the group consisting of TRA F 6, P axil 1 in, Z yxin and J ade-1.
〔33〕 亜鉛イオン濃度の制御が亜鉛イオントランスポーターの発現及び 又は 機能を調節することによって行われるものである、 上記 〔26〕 記載の方法。 [33] The method according to [26] above, wherein the zinc ion concentration is controlled by adjusting the expression and / or function of a zinc ion transporter.
〔34〕 亜鉛イオントランスポーターが L I Vフアミリーを含むヒ ト Z I P類、 ヒ ト CD F類からなる群より選択される少なくとも 1種である、 上記 〔33〕 記 載の方法。 [34] The method according to [33] above, wherein the zinc ion transporter is at least one selected from the group consisting of human Z IPs and human CD Fs containing L IV family.
〔35〕 抗原提示細胞が樹状細胞、 マクロファージ、 ランゲルハンス細胞、 B細 胞、 胸腺上皮細胞、 滑膜細胞、 血管内皮細胞及びケラチノサイ トからなる群より 選ばれる少なくとも 1種である、 上記 〔2 6〕 〜 〔3 4〕 のいずれかに記載の方 法。 [35] From the group consisting of dendritic cells, macrophages, Langerhans cells, B cells, thymic epithelial cells, synovial cells, vascular endothelial cells and keratinocytes. The method according to any one of [2 6] to [3 4] above, which is at least one selected.
〔3 6〕 細胞内の亜鉛イオン濃度を測定することを含む、 抗原提示細胞の機能を 制御し得る化合物のスクリーニング方法。  [36] A method for screening a compound capable of controlling the function of antigen-presenting cells, comprising measuring intracellular zinc ion concentration.
〔3 7〕 抗原提示細胞が樹状細胞、 マクロファージ、 ランゲルハンス細胞、 B細 胞、 胸腺上皮細胞、 滑膜細胞、 血管内皮細胞及びケラチノサイ トからなる群より 選ばれる少なくとも 1種である、 上記 〔3 6〕 記載の方法。  [37] The antigen presenting cell is at least one selected from the group consisting of dendritic cells, macrophages, Langerhans cells, B cells, thymic epithelial cells, synovial cells, vascular endothelial cells, and keratinocytes, [3 6] The method described.
〔3 8〕 抗原提示細胞が樹状細胞である、 上記 〔3 7〕 記載の方法。  [3 8] The method described in [3 7] above, wherein the antigen-presenting cell is a dendritic cell.
〔3 9〕 抗原提示細胞の機能制御剤を製造するための、 細胞内の亜鉛イオン濃度 を制御し得る物質の使用。  [39] Use of a substance capable of controlling the intracellular zinc ion concentration for producing a function control agent for antigen-presenting cells.
〔4 0〕免疫システムが関与する疾患の予防 ·治療薬を製造するための、上記〔1〕 〜 〔1 1〕 いずれかに記載の機能制御剤の使用。  [40] Use of the function-controlling agent according to any one of [1] to [11] above, for producing a prophylactic / therapeutic agent for a disease involving the immune system.
〔4 1〕 免疫システムが関与する疾患の予防 ·治療薬を製造するための、 細胞内 の亜鉛イオン濃度を制御し得る物質の使用。  [4 1] Use of a substance capable of controlling the intracellular zinc ion concentration for the manufacture of a drug for the prevention and treatment of diseases involving the immune system.
図面の簡単な説明  Brief Description of Drawings
図 1は、 樹状細胞を亜鉛イオンキレーターで処理すると、 その細胞表面の MH Cクラス I、 クラス I I、 C D 8 6分子の発現が増強することを示す図である。 図' 2は、 樹状細胞を亜鉛イオン及び亜鉛ィオノフォアで処理すると、 L P S刺 激による樹状細胞の MH C I I及び C D 8 6分子の発現増強や I L— 1 2の産生 が抑制されることを示す図である。  FIG. 1 shows that when dendritic cells are treated with a zinc ion chelator, the expression of MH C class I, class I I, and CD 86 molecules on the cell surface is enhanced. Fig. 2 shows that when dendritic cells are treated with zinc ions and zinc ionophores, LPS-stimulated dendritic cells enhance the expression of MH CII and CD86 molecules and suppress IL-12 production. FIG.
図 3は、 レトロウィルスベクターを用いて m L i v 1を発現させた樹状細胞で は、 L P S刺激による M H Cクラス I I分子の発現増強が抑制されることを示す 図である。  FIG. 3 is a graph showing that dendritic cells in which m L i v 1 is expressed using a retroviral vector suppress the enhancement of MHC class I I molecule expression by L PS stimulation.
発明を実施するための最良の形態  BEST MODE FOR CARRYING OUT THE INVENTION
本発明において「亜鉛イオン濃度の制御」とは亜鉛イオンの細胞内での濃度(量) を増加させる、 あるいは減少させるという文言通りの意味に加え、 最終的に細胞 内で亜鉛イオン癉度が増減することによって生じる現象と同様な現象を誘導する ことができる作用をも意味し、 そのような場合には細胞内での亜鉛イオンめ濃度 の多少には拘束されない。 本発明において、 亜鉛イオン濃度の制御の対象となる 細胞は、 樹状細胞、 マクロファージ、 ランゲ/レハンス細胞、 B細胞、 胸腺上皮細 胞等の抗原提示細胞、 さらには滑膜細胞、 血管内皮細胞、 ケラチノサイ ト等の抗 原提示能を獲得することが予想される細胞である。 好ましくは樹状細胞である。 本明細書では制御の対象となる細胞を便宜上 「抗原提示細胞」 と称することもあ る。 ' In the present invention, “control of zinc ion concentration” means in addition to the literal meaning of increasing or decreasing the concentration (amount) of zinc ion in the cell, and finally the zinc ion concentration increases or decreases in the cell. Induces a phenomenon similar to that caused by In such a case, it is not restricted by the concentration of zinc ions in the cell. In the present invention, the cells to be controlled for zinc ion concentration include dendritic cells, macrophages, Lange / rehans cells, B cells, thymic epithelial cells and other antigen-presenting cells, as well as synovial cells, vascular endothelial cells, These cells are expected to acquire the ability to present antigens such as keratinocytes. Dendritic cells are preferred. In the present specification, the cells to be controlled are sometimes referred to as “antigen-presenting cells” for convenience. '
「細胞内の亜鉛イオン濃度を制御し得る物質」 としては 「亜鉛イオン」、 「亜鉛 イオンキレーター」、 「亜鉛ィオノフォア」、 「亜鉛イオン要求性タンパク質の発現 及び Z又は機能を調節する物質」、 「亜鉛イオントランスポーターの発現及び 又 は機能を調節する物質」等が挙げられるが、 「亜鉛イオン Jや「亜鉛ィオノフォア」 はそれを添加することにより抗原提示細胞内の亜鉛ィオン濃度を直接増加させる ことができ、一方、 「亜鉛イオンキレーター」はそれを添加することにより抗原提 示細胞内の亜鉛イオン濃度を直接低減することができ、 それによつて抗原提示細 胞の機能を制御することができる。 「亜鉛イオン要求性タンパク質の発現及び κ 又は機能を調節する物質」 や 「亜鉛イオントランスポーターの発現及び/又は機 能を調節する物質」 は、 亜鉛イオン要求性タンパク質の発現及び ζ又は機能を調 節することによって、 あるいは亜鉛イオントランスポーターの発現及び ζ又は機 能を調節することによって、 亜鉛イオンの抗原提示細胞に及ぼす作用に変化を生 じさせて抗原提示細胞の機能を制御することができる。 「亜鉛イオン要求性タン パク質の発現及びノ又は機能を調節する物質」 あるいは 「亜鉛イオントランスポ 一ターの発現及び 又は機能を調節する物質」 において、 発現あるいは機能を促 進する方向に調節する物質であれば当該細胞の亜鉛イオンに対する応答性をより 高め、 発現あるいは機能を抑制する方向に調節する物質であれば当該細胞の亜鉛 イオンに対する応答性をより抑制することができる。 本発明において亜鉛イオン 濃度の 「制御」 とは、 正及び負のいずれの調節をも意味する。  “Substances that can control intracellular zinc ion concentration” include “zinc ions”, “zinc ion chelators”, “zinc ionophores”, “substances that regulate the expression and Z or function of zinc ion-requiring proteins”, “ Examples include substances that regulate the expression and / or function of zinc ion transporters, etc.``Zinc ion J or zinc ionophore '' can directly increase the concentration of zinc ions in antigen-presenting cells by adding them. On the other hand, the “zinc ion chelator” can directly reduce the zinc ion concentration in the antigen-presenting cell by adding it, thereby controlling the function of the antigen-presenting cell. “Substances that regulate the expression and κ or function of zinc ion-requiring protein” and “Substances that regulate the expression and / or function of zinc ion transporter” regulate the expression and ζ or function of zinc ion-requiring protein. Or by regulating the expression and ζ or function of the zinc ion transporter, it is possible to control the function of antigen-presenting cells by changing the effect of zinc ions on antigen-presenting cells. . Substance that regulates the expression or function in a direction that promotes the expression or function of a substance that regulates the expression and function of a zinc ion-requiring protein or a substance that regulates the expression and / or function of a zinc ion transporter Then, the responsiveness to the zinc ion of the cell can be further enhanced, and the responsiveness to the zinc ion of the cell can be further suppressed as long as it is a substance that regulates the expression or function. In the present invention, “control” of the zinc ion concentration means both positive and negative regulation.
抗原提示細胞は、 1 ) 感染等に由来する外来物質 (異物)、 並びに 2 ) 自己若し くは細胞の腫瘍化、 ウィルス感染等に起因する内在性タンパク質を TCRを持つ 細胞 (例えば CD4 + T細胞、 CD8+T細胞、 DNT細胞、 NKT細胞等) に提 示する。 そこで自己、 非自己の区別により感染防御や自己にとって好ましくない 細胞の除去を行う。 本発明では、 これらの抗原提示細胞の機能を 「制御する」 こ とにより、 生命の維持にとってより好ましい免疫応答を惹起する。 抗原提示細胞 の機能を 「制御する」 こととは、 より具体的には抗原提示後の免疫応答の質と量 を規定することを意味する。 ここでの 「質」 とは、 CD 4+T細胞において T h 1と T h 2細胞の応答制御と免疫寛容等、 又 CD 8 + T細胞においては抗原特異 的細胞障害活性を持つ C T Lの応答制御と免疫寛容等を意味し、 「量 Jとは文言通 り免疫応答め強さを意味する。又、抗原提示細胞の機能を「制御する」 こととは、 抗原提示細胞そのものの活性化若しくは不活性化を意味する。 具体的には、 1) MHCクラス I、 I I分子の発現制御、 2) CD 80、 CD86、 CD 40等の 副刺激分子の発現制御、 3) I L— 1、 I L— 6、 I L— 1 0、 I L— 1 2、 I FN- a/j3 , TNF_a、 T G F— 等のサイ ト力インの発現制御、 4 ) 外来 若しくは内在性抗原の取り込み制御、 5) 抗原提示細胞の遊走能の制御等から選 ばれる 1以上の項目の機能を制御することを意味する。 Antigen-presenting cells consist of 1) foreign substances (foreign substances) derived from infection, etc., and 2) self Alternatively, endogenous proteins resulting from cell tumorigenesis, virus infection, etc. should be presented to cells with TCR (eg CD4 + T cells, CD8 + T cells, DNT cells, NKT cells, etc.). Therefore, we will protect against infection and remove cells that are undesirable for ourselves by distinguishing between self and non-self. In the present invention, by controlling the function of these antigen-presenting cells, an immune response that is more favorable for the maintenance of life is elicited. “Controlling” the function of antigen-presenting cells means more specifically defining the quality and quantity of immune responses after antigen presentation. “Quality” here means response control and immune tolerance of T h 1 and T h 2 cells in CD 4+ T cells, and response of CTL with antigen-specific cytotoxic activity in CD 8 + T cells. It means control and immune tolerance, etc. “Amount J literally means strength of immune response. Also,“ controlling ”the function of an antigen-presenting cell means activation of the antigen-presenting cell itself or It means inactivation. Specifically, 1) expression control of MHC class I and II molecules, 2) expression control of costimulatory molecules such as CD 80, CD86, and CD 40, 3) IL-1, IL-6, IL-10, IL-1 2, IFN-a / j3, TNF_a, TGF—, etc., control of the expression of site force-in, 4) control of uptake of foreign or endogenous antigen, 5) control of migration ability of antigen-presenting cells, etc. Means to control the function of one or more items.
「亜鉛イオン」 は亜鉛ィオノフォアによって抗原提示細胞内に導入される。 す なわち亜鉛イオンは亜鉛ィオノフォアとの錯体の形で抗原提示細胞内に導入され る。 亜鉛ィオノフォアとしては当分野で通常用いられている、 好ましくは市販さ れている種々の化合物が挙げられる。 亜鉛ピリチオン、 複素環ァミン、 ジチォ力 ルバメート、 ビタミン類等が例示される。  “Zinc ions” are introduced into antigen-presenting cells by zinc ionophores. In other words, zinc ions are introduced into antigen-presenting cells in the form of a complex with a zinc ionophore. Zinc ionophores include various compounds that are commonly used in the art, and preferably are commercially available. Examples include zinc pyrithione, heterocyclic amine, dithiol rubamate, and vitamins.
「亜鉛イオンキレーター」 とは、 抗原提示細胞内で亜鉛イオンと錯体を形成す ることによって細胞内から亜鉛イオンを除去することができる物質であって、 例 えば、 2 , 3—ジメルカプト― 1一プロパンスルホン酸(DMP S)、 N, N, Ν', N' —テ トラキス (2—ピリジルメチル) エチレンジァミン (ΤΡΕΝ)、 ェチレ ンジァミン (EDTA) 及び Ν— (6—メ トキシ一 8—キノ リル) 一 ρ— トルェ ンスルホンアミ ド(T SQ)等が挙げられる。いずれも商業的に入手可能である。 また、 本発明の抗原提示細胞の機能制御剤、 本発明の免疫システムが関与する疾 患の予防 ·治療薬 (以下、 単に予防 ·治療薬と称することもある) に有効成分と して含められる亜鉛イオン濃度を制御し得る物質としての亜鉛イオンキレーター の量は、 それぞれ細胞内での亜鉛イオン濃度の制御が可能な範囲で適宜決定され る。 また、 含める亜鉛イオンキレータ一は 1種類であっても 2種類以上であって もよい。 2種類以上含める場合にはその種類に応じて配合量を適宜設定する。 A “zinc ion chelator” is a substance that can remove zinc ions from cells by forming a complex with zinc ions in antigen-presenting cells. For example, 2, 3-dimercapto-1 Propanesulfonic acid (DMP S), N, N, Ν ', N' — Tetrakis (2-pyridylmethyl) Ethylenediamine (ΤΡΕΝ), Ethylenediamine (EDTA) and Ν— (6-Methoxy 8-quinolyl) Ρ-Toluenesulfonamide (T SQ) and the like. Both are commercially available. In addition, it is included as an active ingredient in the function-controlling agent of the antigen-presenting cell of the present invention, and a prophylactic / therapeutic drug for a disease involving the immune system of the present invention (hereinafter sometimes simply referred to as a prophylactic / therapeutic drug). The amount of zinc ion chelator as a substance capable of controlling the zinc ion concentration is appropriately determined within a range in which the intracellular zinc ion concentration can be controlled. One zinc ion chelator may be included, or two or more zinc ion chelators may be included. When two or more types are included, the blending amount is appropriately set according to the type.
「亜鉛イオン要求性タンパク質の発現及び Z又は機能を調節する物質」 とは亜 鉛イオン要求性タンパク質の発現及び 又は機能を結果的に調節することができ ればその作用機序は特に限定されず、 遺伝子レベルでの調節であってもタンパク 質レベルでの調節であってもよい。 遺伝子レベルでの調節としては、 転写調節、 遺伝子発現調節等が挙げられる。タンパク質レベルでの調節としては、代謝調節、 リン酸化、 脱リン酸化、 糖付加、 脂質付加、 亜鉛との配位結合、 分解、 ュビキチ ン化、 ァセチル化等が挙げられる。 「亜鉛イオン要求性タンパク質」 は、抗原提示 細胞の機能発現に亜鉛が必要なタンパク質であって、 例えば亜鉛フィンガーを有 するタンパク質、 R I NGフィンガーを有するタンパク質、 L I Mドメインを有 するタンパク質、 PHD亜船フィンガーを有するタンパク質等が挙げられる。 亜 鉛フィンガーは、 システィンゃヒスチジンといったアミノ酸が亜鉛と配位するこ とによって初めて高次構造を有することができるユニークな核酸結合能を有する モチーフであり、 亜鉛フィンガーを有するタンパク質としては、 例えば、 上皮一 間葉転換と呼ばれる現象(ヒ トやその他の生物の初期発生における体の形作りや、 傷口の治癒 ·癌の転移等の際に、 通常密に結合している細胞同士が隣の細胞との 接着を解除し可動性を獲得し他へ移動する現象) のマスターレギュレーターであ る転写因子 S n a i 1、 PKC a、 GEF— H l、 HDAC、 SQSTM1等が 例示される。 R I NGフィンガーは、 亜鉛フィンガーの変形ともいえ、 タンパク 質間の相互作用に関与しているものと示唆されており、 R I NGフィンガーを有 するタンパク質としては、 ュビキチン一タンパク質リガーゼ E 3 (u b i q u i t i n-p r o t e i n l i g a s e E 3 ) 活性を有するものや、 ュビキチ ン共役タンノ ク質 E 2 (u b i q u i t i n— c o n j u g a t i n g e n z yme s E 2)、 TRAF 6 (T N F αのシグナル伝達分子) との結合を示すも のが報告されている。 L IMドメインは、 6つのシスティンと 1つのヒスチジン の位置が保存された 60アミノ酸からなり、 亜鉛フィンガーとよく似た構造をと るものの DNA結合能は知られていない。 PKCとの結合に介在するドメインと しても機能する。 L I Mドメインを有するタンパク質としては、 細胞骨格制御に 関与する P a'x i 1 1 i n、 Z y x i n等が例示される。 PHD亜鉛フィンガー は、 クロマチンを媒介した転写調節に関与することが示唆ざれる核タンパクであ る亜鉛フィンガ一様ドメインであり、 DNA結合能を有するものと予測されてい る。 PHD亜鉛フィンガーを有するタンパク質としては、 ヒス トンのァセチル化 に関与する J a d e— 1等が例示される。 “Substances that regulate the expression and Z or function of zinc ion-requiring protein” are not particularly limited as long as the expression and / or function of zinc ion-requiring protein can be regulated as a result. It may be regulation at the gene level or regulation at the protein level. Examples of regulation at the gene level include transcription regulation and gene expression regulation. Examples of regulation at the protein level include metabolic regulation, phosphorylation, dephosphorylation, sugar addition, lipid addition, coordinate bond with zinc, degradation, ubiquitination, acetylation, and the like. “Zinc ion-requiring protein” is a protein that requires zinc for the functional expression of antigen-presenting cells. For example, a protein having a zinc finger, a protein having a RING finger, a protein having a LIM domain, or PHD Examples include proteins having fingers. A zinc finger is a unique nucleic acid-binding motif that can have a higher-order structure only when an amino acid such as cystine-histidine is coordinated with zinc. Examples of proteins having a zinc finger include epithelium. A phenomenon called mesenchymal transition (such as body shaping in the early development of humans and other organisms, wound healing, cancer metastasis, etc.) Examples include transcription factors Snai1, PKCa, GEF—Hl, HDAC, SQSTM1, etc., which are master regulators of the phenomenon of releasing adhesion, acquiring mobility and moving to another). The RI NG finger is a variant of the zinc finger, and is suggested to be involved in protein-protein interactions, and ubiquitin monoprotein ligase E 3 (ubiquitin np roteinligase) is a protein with a RI NG finger. E 3) Active or ubiquitous It has been reported that it binds to conjugated nucleoprotein E 2 (ubiquitin—conjugating enzymes E 2) and TRAF 6 (TNF α signaling molecule). The L IM domain consists of 60 amino acids in which the positions of 6 cysteines and 1 histidine are conserved. Although it has a structure similar to that of zinc fingers, its DNA-binding ability is unknown. It also functions as a domain that mediates binding to PKC. Examples of proteins having a LIM domain include Pa'x i 11 in, Z yxin, etc. involved in cytoskeletal regulation. The PHD zinc finger is a zinc finger uniform domain, a nuclear protein that is implicated in chromatin-mediated transcriptional regulation, and is predicted to have DNA-binding ability. Examples of the protein having a PHD zinc finger include Jade-1 involved in histone acetylation.
「亜鉛イオン要求性タンパク質の発現及びノ又は機能を調節する物質」 は、 公 知の化合物であっても今後開発される新規な化合物であってもよい。 また、 低分 子化合物であっても高分子化合物であってもかまわない。 ここで低分子化合物と は分子量 3000未満程度の化合物であって、 例えば医薬品として通常使用し得 る有機化合物及びその誘導体や無機化合物が挙げられ、 有機合成法等を駆使して 製造される化合物やその誘導体、 天然由来の化合物やその誘導体、 プロモーター 等の小さな核酸分子や各種の金属等であり、 望ましくは医薬品として使用し得る 有機化合物及びその誘導体、 核酸分子をいう。 また、 高分子化合物としては分子 量 3000以上程度の化合物であって、 タンパク質、 ポリ核酸類、 多糖類、 及び これらを組み合わせたもの等が挙げられ、 望ましくはタンパク質である。 これら の低分子化合物あるいは高分子化合物は、 公知のものであれば商業的に入手可能 であるか、 各報告文献に従って採取、 製造、 精製等の工程を経て得ることができ る。 これらは、 天然由来であっても、 また遺伝子工学的に調製されるものであつ てもよく、 また半合成等によっても得ることができる。 具体的には、 亜鉛イオン 要求性タンパク質 S n a i 1は、 T G F— ]3又は F G Fを通して M A P Kにより その発現が調節される (Peinado, H. , Quintanilla, . & Cano, A. Transforming growth factor beta - 1 induces snail transcription factor in epithelial cell lines : mechanisms for epithelial mesenchymal transitions. J. Biol. Chem. 278, 21113-23 (2003))。 また、 S n a i 1は亜鉛イオントランスポーターである L I V 1 (後述)により活性化される(Yamashita, S. , Miyagi, C. , Fukada, Τ· , Kagara, Ν. , Che, Υ, - S. & Hirano, Τ. Zinc transporter LIVI controls The “substance that regulates the expression and function of zinc ion-requiring protein” may be a known compound or a new compound that will be developed in the future. Further, it may be a low molecular compound or a high molecular compound. Here, the low molecular weight compound is a compound having a molecular weight of less than about 3000, and includes, for example, organic compounds that can be normally used as pharmaceuticals and their derivatives and inorganic compounds. Derivatives, naturally-occurring compounds and derivatives thereof, small nucleic acid molecules such as promoters, various metals, and the like, desirably organic compounds that can be used as pharmaceuticals, derivatives thereof, and nucleic acid molecules. The polymer compound is a compound having a molecular weight of about 3000 or more, and includes proteins, polynucleic acids, polysaccharides, and combinations thereof, and is preferably a protein. These low-molecular compounds or high-molecular compounds are commercially available if they are known, or can be obtained through steps such as collection, production, and purification according to each report literature. These may be of natural origin, prepared by genetic engineering, or obtained by semisynthesis. Specifically, the expression of zinc auxotrophic protein S nai 1 is regulated by MAPK through TGF-] 3 or FGF (Peinado, H., Quintanilla,. & Cano, A. Transforming growth factor beta-1 induces snail transcription factor in epithelial cell lines: mechanisms for epithelial mesenchymal transitions. J. Biol. Chem. 278, 21113-23 (2003)). Snai 1 is activated by LIV 1 (described later), which is a zinc ion transporter (Yamashita, S., Miyagi, C., Fukada, Τ ·, Kagara, Ν., Che, Υ, -S. & Hirano, Τ. Zinc transporter LIVI controls
epithelial- mesenchymal transition in zebraf ish gastru丄 a organizer. Nature 429, 298 - 302 (2004))。 他の亜鉛イオン要求性タンパク質として例示される R a f 1、 GEF— Η ί、 PKCひ、 フォスファターゼ、 メタ口チォネインについて はそれてれ Jirakulaporn T, Muslin AJ. Cation diffusion racilitator proteirrs modulate Raf-1 activity. J. Biol. Chem. 279, 27807 - 15 (2004) NKrendel M, Zenke FT, Bokoch GM. Nucleotide exchange factor GEF - HI mediates cross-talk between microtubules and the actin cytoskeleton. Nature Cell Biology 4, 294 - 301 (2002)、 Korichneva I, Hoyos B, し hua R, Levi E, Hammer ling U. Zinc release from protein kinase C as the common event during activation by lipid second messenger or reactive oxygen. J. Biol. Chem. 277, 44327-31 (2002)、 Haase H, Maret W. Intracellular zinc fluctuations modulate protein tyrosine phosphatase activity in insulin/insulin-like growth factor - 1 signaling. Exp. Cell Research 291, 289-298 (2003)、 生化学辞典 (第 3版), (株) 東京化学同人 発行, 第 1 393頁 ( 1 998) に詳細が記載されている。 TRAF 6、 P a x i l l i n、 Z y x i n、 J a d e— 1についてはそれぞれ Kobayashi , Kadono Y, Naito A, Matsumoto K, Yamamoto T, Tanaka S, Inoue J. Segregation of TRAF6 - mediated signaling pathways clarities its role in osteoclastogenesis. The EMB0 J. 20, 1271-1280 (2001)、 Brown MC, Perrotta JA, Turner CE. epithelial-mesenchymal transition in zebraf ish gastru 丄 a organizer. Nature 429, 298-302 (2004)). For other zinc ion-requiring proteins, Raf 1, GEF— Η ί, PKC, phosphatase, and meta-mouthonein are also available. Jirakulaporn T, Muslin AJ. Cation diffusion racilitator proteirrs modulate Raf-1 activity. J Biol. Chem. 279, 27807-15 (2004) N Krendel M, Zenke FT, Bokoch GM.Nucleotide exchange factor GEF-HI mediates cross-talk between microtubules and the actin cytoskeleton.Nature Cell Biology 4, 294-301 (2002) ), Korichneva I, Hoyos B, Shihua R, Levi E, Hammer ling U. Zinc release from protein kinase C as the common event during activation by lipid second messenger or reactive oxygen. J. Biol. Chem. 277, 44327-31 (2002), Haase H, Maret W. Intracellular zinc fluctuations modulate protein tyrosine phosphatase activity in insulin / insulin-like growth factor-1 signaling. Exp. Cell Research 291, 289-298 (2003), Biochemical Dictionary (3rd edition) ), Published by Tokyo Chemical Doujin, page 1 393 (1 998) for details. For TRAF 6, P axillin, Z yxin, J ade— 1 Kobayashi, Kadono Y, Naito A, Matsumoto K, Yamamoto T, Tanaka S, Inoue J. Segregation of TRAF6-mediated signaling pathways clarities its role in osteoclastogenesis. EMB0 J. 20, 1271-1280 (2001), Brown MC, Perrotta JA, Turner CE.
Identification of LIM3 as the principal determinant of paxillin focal adhesion localization and characterization of a novel motif on paxillin directing vinculin and focal adhesion kinase binding. J. Cell Biol. 135, 1109-1123 (1996)、 Sadler I, Crawford AW, Michelsen JW, Beckerle MC. Zyxin and cCRP: two interactive LIM domain proteins associated with the cytoskeleton. J. Cell Biol. 119, 1573 - 1587 (1992)、 Panchenko MV, Zhou MI, Cohen HT. von Hippel-Lindau partner Jade - 1 is a transcriptional co-activator associated with histone acetyl transferase activity. J. Biol. Chem. 279, 56032-56041 (2004)に詳細が記載されている。 さらに S Q S TM 1及び HD A C こつレヽ飞 i¾てれてれ Joung I, Strominger JL, Shin J. Molecular cloning of a phosphotyrosine - independent ligand of the p561ck SH2 domain. Proc Natl Acad Sci USA, 93, 5991-5995 (1996)及び Kawaguchi Y, Kovacs JJ, McLaurin A, Vance JM, Ito A, Yao TP. The deacetylase HDAC6 regulates aggresome formation and cell viability in response to misfolded protein stress. Cell 115, 727 - 738 (2003)に詳細が記載されている。 Identification of LIM3 as the principal determinant of paxillin focal adhesion localization and characterization of a novel motif on paxillin directing vinculin and focal adhesion kinase binding.J. Cell Biol. 135, 1109-1123 (1996), Sadler I, Crawford AW, Michelsen JW , Beckerle MC. Zyxin and cCRP: two interactive LIM domain proteins associated with the cytoskeleton.J. Cell Biol.119, 1573-1587 (1992), Panchenko MV, Zhou MI, Cohen HT.von Hippel-Lindau partner Jade-1 is a transcriptional co-activator details are described in associated with histone acetyl transferase activity. J. Biol. Chem. 279, 56032-56041 (2004). In addition, SQS TM 1 and HD AC can be used as an example Joung I, Strominger JL, Shin J. Molecular cloning of a phosphotyrosine-independent ligand of the p561ck SH2 domain. Proc Natl Acad Sci USA, 93, 5991-5995 ( 1996) and Kawaguchi Y, Kovacs JJ, McLaurin A, Vance JM, Ito A, Yao TP.The deacetylase HDAC6 regulates aggresome formation and cell viability in response to misfolded protein stress.Cell 115, 727-738 (2003). Has been.
「亜鉛イオントランスポーターの発現及び 又は機能を調節する物質」 とは亜 鉛イオントランスポーターの発現及び Z又は機能を結果的に調節することができ ればその作用機序は特に限定されず、 遺伝子レベルでの調節であってもタンパク 質レベルでの調節であってもよい。 遺伝子レベルでの調節としては、 転写調節、 遺伝子発現調節等が挙げられる。タンパク質レベルでの調節としては、代謝調節、 リン酸化、 脱リン酸化、 糖付加、 脂質付加、 亜鉛との配位結合、 分解、 ュビキチ ン化、 ァセチル化等が挙げられる。 「亜鉛イオントランスポーター」 としては、 例 えば、 L I Vフアミ リーを含むヒ ト Z I P類 (BAB 70848、 h Z i p 4、 B I GM1 03、 K I AA006 2、 K I AA 1 265、 h L i v— 1、 A AH 088 53、 hKE 4、 XP— 20864 9、 h Z I P l、 h Z I P 2、 h Z I P 3、 BAA 9 2 1 00, BAC04504等)、 ヒ ト CDF類 (h Z n t _ 5、 h Z n t— 7、 h Z n t— 1、 h Z n t— 6、 h Z n t— 3、 h Z n t— 2、 h Z n t— 8、 h Z n t— 4、 h Z n t— 9等) 等が挙げられる。 L I V 1は、 当 初はエス トロゲン制御を受ける乳がんタンパク質として同定され、 最近になって LZT (Z I P亜鉛イオン卜ランスポーターの L I V 1サブファミリー) と称さ れる Z I P亜鉛イオントランスポーターサブファミリ一 (Z r t―、 I r t様タ ンパク質) に属し (Taylor, K. M. & Nicholson, R. I. The LZT proteins ; the LIV-1 subfami ly of zinc transporters. Biochim. Biophys. Acta 1611, 16 - 30 (2003) )、 亜船イオントランスポーターとして機能することが明らかになった (Taylor, K. ., Morgan, H. E., Johnson, A., Hadley, し J. & Nicholson, R. I. “A substance that regulates the expression and / or function of zinc ion transporter” means that the mechanism of action is not particularly limited as long as the expression and Z or function of zinc ion transporter can be regulated as a result. It may be at the level or at the protein level. Examples of regulation at the gene level include transcription regulation and gene expression regulation. Examples of regulation at the protein level include metabolic regulation, phosphorylation, dephosphorylation, sugar addition, lipid addition, coordinate bond with zinc, degradation, ubiquitination, acetylation, and the like. “Zinc ion transporters” include, for example, human ZIPs containing LIV family (BAB 70848, h Z ip 4, BI GM1 03, KI AA006 2, KI AA1 265, h L iv—1, A AH 088 53, hKE 4, XP— 20864 9, h ZIP l, h ZIP 2, h ZIP 3, BAA 9 2 100, BAC04504, etc., human CDFs (h Z nt _ 5, h Z nt— 7 HZnt-1, 1, hZnt-6, hZnt-3, hZnt-8, hZnt-4, hZnt-9, etc.). LIV 1 was originally identified as an estrogen-controlled breast cancer protein, and recently the Zinc ion transporter subfamily (Z rt) called LZT (LIV 1 subfamily of ZIP zinc ion transporters). ― I rt (Taylor, KM & Nicholson, RI The LZT proteins; the LIV-1 subfamily of zinc transporters. Biochim. Biophys. Acta 1611, 16-30 (2003)) and function as a sub-ion ion transporter (Taylor, K.., Morgan, HE, Johnson, A., Hadley, Shi J. & Nicholson, RI
Structure-function analysis of LIV-1, tne breast cancer-associated protein that belongs to a new subfamily of zinc transporters. Biochem. J. 375, 51 - 9 (2003) )。他 C D F ( c a t i o n d i f f u s i o n f a c i 1 i t a t o r )等が挙げられる。例えば、亜鉛イオントランスポーターである L I V 1は、 S T A T 3により発現調節を受ける (Yamashita, S., iyagi, C. , Fukada, Τ.', Kagara, N. , Che, Y. - S. & Hirano, T. Zinc transporter LIVI controls epithelial - mesenchymal transition in zebraf ish gastrula organizer. Nature 429, 298-302 (2004) )。 Structure-function analysis of LIV-1, tne breast cancer-associated protein that belongs to a new subfamily of zinc transporters. Biochem. J. 375, 51-9 (2003)). Other examples include C DF (c a t i o n d i f f u s i o nf a c i 1 i tat o r) and the like. For example, the zinc ion transporter LIV 1 is regulated by STAT 3 (Yamashita, S., iyagi, C., Fukada, Τ. ', Kagara, N., Che, Y.-S. & Hirano , T. Zinc transporter LIVI controls epithelial-mesenchymal transition in zebraf ish gastrula organizer. Nature 429, 298-302 (2004)).
「亜鉛イオントランスポーターの発現及びノ又は機能を調節する物質」 は、 公 知の化合物であっても今後開発される新規な化合物であってもよい。 また、 低分 子化合物であっても高分子化合物であってもかまわない。 ここで低分子化合物と は分子量 3 0 0 0未満程度の化合物であって、 例えば医薬品として通常使用し得 る有機化合物及びその誘導体や無機化合物が挙げられ、 有機合成法等を駆使して 製造される化合物やその誘導体、 天然由来の化合物やその誘導体、 プロモーター 等の小さな核酸分子や各種の金属等であり、 望ましくは医薬品として使用し得る 有機化合物及びその誘導体、 核酸分子をいう。 また、 高分子化合物としては分子 量 3 0 0 0以上程度の化合物であって、 タンパク質、 ポリ核酸類、 多糖類、 及び これらを組み合わせたもの等が挙げられ、 望ましくはタンパク質である。 これら の低分子化合物あるいは高分子化合物は、 公知のものであれば商業的に入手可能 であるか、 各報告文献に従って採取、 製造、 精製等の工程を経て得ることができ る。 これらは、 天然由来であっても、 また遺伝子工学的に調製されるものであつ てもよく、 また半合成等によっても得ることができる。  The “substance that regulates the expression and function of zinc ion transporter” may be a known compound or a new compound to be developed in the future. Further, it may be a low molecular compound or a high molecular compound. Here, the low molecular weight compound is a compound having a molecular weight of less than about 300, for example, an organic compound that can be usually used as a pharmaceutical, a derivative thereof, and an inorganic compound. Compounds, derivatives thereof, naturally derived compounds, derivatives thereof, small nucleic acid molecules such as promoters and various metals, and preferably organic compounds that can be used as pharmaceuticals, derivatives thereof, and nucleic acid molecules. The polymer compound is a compound having a molecular weight of about 300 or more, and includes proteins, polynucleic acids, polysaccharides, and combinations thereof, and is preferably a protein. These low-molecular compounds or high-molecular compounds are commercially available if they are known, or can be obtained through steps such as collection, production, and purification according to each report literature. These may be of natural origin, prepared by genetic engineering, or obtained by semisynthesis.
亜鉛イオントランスポーターである L I V 1は亜鉛イオン要求性タンパク質で W LIV 1, a zinc ion transporter, is a zinc ion-requiring protein W
14 ある S n a i 1の活性(特にレブレッサー活性)を増強することから(Yama^hita, S., Miyagi, C. , Fukada, X. , Kagara, N. , Che, Y.— S. & Hirano, T. Zinc transporter LIVI controls epithelial - mesenchymal transition in zebraf ish gastrula organizer. Nature 429, 298-302 (2004))、 本発明における 「亜鋭ィォ ン要求性タンパク質の発現及び Z又は機能を調節する物質」 の一例として以下の ものが挙げられる。  14 Because it enhances the activity of S nai 1 (especially repressor activity) (Yama ^ hita, S., Miyagi, C., Fukada, X., Kagara, N., Che, Y.— S. & Hirano, T. Zinc transporter LIVI controls epithelial-mesenchymal transition in zebraf ish gastrula organizer.Nature 429, 298-302 (2004)), `` Substances that regulate the expression and Z or function of subthin requirement protein '' in the present invention The following are examples.
(1) 下記 a) 〜d) のいずれかに記載の DN A  (1) DN A as described in any of a) to d) below
a) 配列番号 2、 4又は 6記載のアミノ酸配列を有するタンパク質 (L I V 1タ ンパク質) をコードする DNA - b) 配列番号 1、 3又は 5記載の配列からなる DNA (L I V 1遺伝子) c) 配列番号 2、 4又は 6記載のアミノ酸配列において 1若しくは複数のァミノ 酸配列が置換、 欠失、 挿入及び Z又は付加されたアミノ酸配列を有するタンパク 質 (L I V 1に類似するタンパク質) をコードした DNA  a) DNA encoding a protein having the amino acid sequence described in SEQ ID NO: 2, 4 or 6 (LIV 1 protein)-b) DNA comprising the sequence described in SEQ ID NO: 1, 3 or 5 (LIV 1 gene) c) DNA encoding a protein (protein similar to LIV 1) having an amino acid sequence in which one or more amino acid sequences are substituted, deleted, inserted and Z or added in the amino acid sequence described in SEQ ID NO: 2, 4 or 6
d ) 配列番号 1、 3又は 5記載の配列とス トリンジェントな条件でハイブリダイ ズする DNA d) DNA that hybridizes with the sequence of SEQ ID NO: 1, 3 or 5 under stringent conditions
(2) 下記 a) 〜d) のいずれかに記載の DNAが挿入されたベクター  (2) A vector into which the DNA according to any one of a) to d) below is inserted
a ) 配列番号 2、 4又は 6記載のアミノ酸配列を有するタンパク質をコードする DNA a) DNA encoding a protein having the amino acid sequence of SEQ ID NO: 2, 4 or 6
b) 配列番号 1、 3又は 5記載の配列からなる DN A b) DN A consisting of the sequence described in SEQ ID NO: 1, 3 or 5
c) 配列番号 2、 4又は 6記載のアミノ酸配列において 1若しくは複数のァミノ 酸配列が置換、 欠失、 揷入及び Z又は付加されたアミノ酸配列を有するタンパク 質をコードした DNA c) DNA encoding a protein having an amino acid sequence in which one or more amino acid sequences are substituted, deleted, inserted and Z or added in the amino acid sequence described in SEQ ID NO: 2, 4 or 6
d ) 配列番号 1、 3又は 5記載の配列とス トリンジェントな条件でハイブリダィ ズする DNA d) DNA that hybridizes with the sequence described in SEQ ID NO: 1, 3 or 5 under stringent conditions
(3) 下記 a) 〜d) のいずれかに記載の DNAによってコードされたタンパク 質  (3) A protein encoded by the DNA according to any one of a) to d) below
a) 配列番号 2、 .4又は 6記載のアミノ酸配列を有するタンパク質をコードする DNA a) encodes a protein having the amino acid sequence of SEQ ID NO: 2, 4 or 6 DNA
b) 配列番号 1、 3又は 5記載の配列からなる DNA b) DNA consisting of the sequence described in SEQ ID NO: 1, 3 or 5
c) 配列番号 2、 4又は 6記載のアミノ酸配列において 1若しくは複数のァミノ 酸配列が置換、 欠失、 挿入及び Z又は付加されたアミノ酸配列を有するタンパク 質をコードした DN A c) DNA encoding a protein having an amino acid sequence in which one or more amino acid sequences are substituted, deleted, inserted and Z or added in the amino acid sequence of SEQ ID NO: 2, 4 or 6
d ) 配列番号 1、 3又は 5記載の配列とストリンジ工ントな条件でハイブリダイ ズする DNA' d) DNA 'that hybridizes with the sequence described in SEQ ID NO: 1, 3 or 5 under stringent conditions.
(4) 配列番号 1、 3又は 5記載の配列からなる DN Aを標的配列としたアンチ センスオリ ゴヌクレオチド  (4) Antisense oligonucleotides whose target sequence is DNA consisting of the sequence described in SEQ ID NO: 1, 3 or 5
(5) 配列番号 1、 3又は 5記載の配列からなる DN Aの一部と同一又は類似す る配列を有する二本鎖 RN A  (5) Double-stranded RNA having a sequence identical or similar to a part of DNA consisting of the sequence described in SEQ ID NO: 1, 3 or 5
(6) 配列番号 2、 4又は 6記載のアミノ酸配列を有するタンパク質に対する抗 体  (6) An antibody against a protein having the amino acid sequence set forth in SEQ ID NO: 2, 4 or 6
(7) 配列番号 2、 4又は 6記載のアミノ酸配列を有するタンパク質と会合し、 その機能を調節する物質 (天然物、 非天然物を含む)  (7) Substances that associate with the protein having the amino acid sequence of SEQ ID NO: 2, 4 or 6 and regulate its function (including natural products and non-natural products)
(1 ) 〜 (3) は亜鉛イオン要求性タンパク質である S n a i 1の発現及ぴ 又は機能を正に調節する (発現を増強する、 及び Z又は機能を活性化する) 物質 であり、 (4) 及び(5) は S n a i 1の発現及び/又は機能を負に調節する (発 現を減少させる、 及び/又は機能を抑制する) 物質である。  (1) to (3) are substances that positively regulate the expression and / or function of Snai 1 which is a zinc ion-requiring protein (enhance expression and activate Z or function) (4 ) And (5) are substances that negatively regulate the expression and / or function of Snai 1 (reduce expression and / or suppress function).
L I V 1タンパク質の調製は、 当業者に周知の各種方法によって行うことが可 能である。 例えば、 配列番号 1、 3又は 5に記載の塩基配列からなる DN A (L I V I遺伝子) が挿入されたベクターを保持した形質転換体にタンパク質を生産 させ、 精製することによって調製できる。 使用するベクターはタンパク質生産に 用いる翻訳系により適宜選択することができる。また L I V 1は、乳房、前立腺、 脳下垂体、 脳等のホルモン系組織に発現することが知られている (Taylor, K. M. & Nicholson, R. 丄. The LZT proteins; the LIV - 1 subfamily of zinc transporters. Biochim. Biophys. Acta 1611, 16-30 (2003))。 抗 L I V 1タンパク質抗体を周 知方法で調製し、 該抗体でァフィ二ティカラムを作製すれば、 L I V 1を発現す る細胞抽出物から L I V 1タンパク質を精製することができる。 The LIV 1 protein can be prepared by various methods well known to those skilled in the art. For example, it can be prepared by producing and purifying a protein in a transformant holding a vector having the DNA (LVI gene) having the nucleotide sequence set forth in SEQ ID NO: 1, 3 or 5 inserted therein. The vector to be used can be appropriately selected depending on the translation system used for protein production. LIV 1 is known to be expressed in hormonal tissues such as breast, prostate, pituitary gland, and brain (Taylor, KM & Nicholson, R. 丄. The LZT proteins; the LIV-1 subfamily of zinc transporters. Biochim. Biophys. Acta 1611, 16-30 (2003)). Around anti-LIV 1 protein antibody LIV 1 protein can be purified from a cell extract expressing LIV 1 by preparing it by an intelligent method and preparing a affinity column with the antibody.
L I V 1に類似するタンパク質は亜鉛イオン要求性タンパク質 (例えば S n a i 1 ) の活性を調節し得るタンパク質であって、 例として、 配列番号 2、 4又は 6に記載のアミノ酸配列において 1若しくは複数のアミノ酸配列が置換、 欠失、 挿入及びノ又は付加されたアミノ酸配列からなるタンパク質、 配列番号 1、 3又 は 5に記載の'塩基配列からなる DNAとストリンジェントな条件下でハイブリダ ィズする DN Aによってコードされるタンパク質を挙げることができる。  A protein similar to LIV 1 is a protein capable of regulating the activity of a zinc ion-requiring protein (for example, S nai 1), for example, one or more amino acids in the amino acid sequence set forth in SEQ ID NO: 2, 4 or 6 DNA that hybridizes under stringent conditions with a protein consisting of an amino acid sequence with substitution, deletion, insertion, and addition or deletion, or with a DNA consisting of a base sequence according to SEQ ID NO: 1, 3 or 5 The protein encoded by can be mentioned.
L I V 1に類似するタンパク質の調製は、 例えば、 L I V Iをコードする塩基 配列を利用してハイブリダィゼーシヨンを行い、 得られた相同性の高い DN Aに よって形質転換体を作製し、 該形質転換体に所望のタンパク質を生産させる方法 をとることができる。相同性の高い DNAを得る方法の一例として、配列番号 1、 3又は 5に記載された塩基配列の一部をプローブとし、 ヒ トゃヒ ト以外の脊椎動 物の細胞等からストリンジェントなハイブリダィゼーション条件下でハイブリダ ィズする方法を挙げることができる。  For the preparation of a protein similar to LIV 1, for example, hybridization is performed using a base sequence encoding LIVI, a transformant is prepared using the obtained highly homologous DNA, A method can be used in which the transformant produces the desired protein. As an example of a method for obtaining highly homologous DNA, a part of the base sequence described in SEQ ID NO: 1, 3 or 5 is used as a probe, and stringent hybrids are obtained from cells of vertebrate animals other than human rabbits. An example is a method of hybridizing under dithering conditions.
上記ストリンジェントなハイブリダィゼーシヨン条件は、 当業者であれば、 適 宜選択することができる。 一例を示せば 25%ホルムアミ ド、 より厳しい条件で は 50%ホルムアミ ド、 4 X S SC、 50mM He p e s pH 7. 0、 1 0 Xデンハルト溶液、 20 8 11 し変性サケ精子0 を含むハィブリダイゼー シヨン溶液中、 42°Cで一晚プレハイブリダィゼーシヨンを行った後、 標識した プローブを添加し、 42 °Cでー晚保温することによりハイプリダイゼーションを 行う。 その後の洗浄における洗浄液及び温度条件は 「 1 X S S C、 0. 1 % S D S、 3 7°C」 程度で、 より厳しい条件としては 「0. 5 X S S C、 0. 1 % S D S、 42°CJ 程度で、 さらに厳しい条件としては 「0. 2 X S SC、 0. 1 %S DS、 65 °C」 程度で実施することができる。 このようにハイブリダィゼーショ ンの洗浄の条件が厳しくなるほどプローブ配列と高い相同性を有する D N Aの単 離を期待し得る。佴し S S C、 SD S及び温度の条件の組み合わせは例示であり、 当業者であれば、 ハイブリダィゼーシヨンのス ト リンジエンシーを決定ずる上記 若しくは他の要素 (例えばプローブ濃度、 プローブの長さ、 ハイブリダィゼーシ ヨン反応時間等) を適宜組み合わせることにより、 上記と同様のス トリンジェン シーを実現することが可能である。 Those skilled in the art can appropriately select the stringent hybridization conditions. For example, a hybrid solution containing 25% formamide, 50% formamide under more severe conditions, 4 XS SC, 50 mM Hepes pH 7.0, 10 X Denhardt's solution, 20 8 11 and denatured salmon sperm 0 After prehybridization at 42 ° C, add labeled probe and incubate at 42 ° C for high prehybridization. In the subsequent cleaning, the cleaning solution and temperature conditions are about `` 1 XSSC, 0.1% SDS, 37 ° C '', and more severe conditions are `` 0.5 XSSC, 0.1% SDS, 42 ° CJ, More severe conditions can be implemented at “0.2 XS SC, 0.1% SDS, 65 ° C”. Thus, isolation of DNA having high homology with the probe sequence can be expected as the conditions for washing hybridization are more severe. The combination of SSC, SD S and temperature conditions is an example, A person skilled in the art can appropriately combine the above or other factors (for example, probe concentration, probe length, hybridization reaction time, etc.) that determine the stringency of the hybridization. It is possible to achieve the same stringency as
このようなハイブリダィゼ一ション技術を利用して単離される DNAがコード するポリペプチドは、 通常、 L I V 1 とアミノ酸配列において高い相同性を有す る。 高い相^性とは、 少なく とも 40%以上、 好ましくは 60%以上、 さらに好 ましくは 80 %以上、 より好ましくは 90 %以上、 より一層好ましくは 95 %以 上、 特に好ましくは 97%以上 (例えば 98~ 9 9%) の配列の相同性を指す。 アミノ酸配列の同一性は、 例えば Ka r 1 i n a n d A l t s c h u lによ るアルゴリズム B LAST (P r o c. N a t l . Ac a d. S c i . USA 8 7 : 2264- 2268, 1 990、 P r o c. Na t l . Ac a d. S c に USA 90 : 587 3- 58 77, 1 9 9 3 )によって決定することができる。 このアルゴリズムに基づいて B LAS TXと呼ばれるプログラムが開発されてい る (A l t s c h u l e t a 1. J . M o 1. B i o l . 2 1 5 : 40 3— 4 10, 1 990)。 B LAS TXによってァミノ酸配列を解析する場合には パラメータ一は例えば s c o r e = 50、 wo r d l e n g t h = 3とする。 B LASTと G a p p e d B L A S Tプログラムを用いる場合には、 各プロダラ ムのデフォルトパラメーターを用いる。 これらの解析方法の具体的な手法は公知 でめる 、h t t p : ZZwww. n c b i . n 1 m. n i h . g o v.)。  A polypeptide encoded by DNA isolated using such a hybridization technique usually has a high homology in amino acid sequence with L I V 1. High compatibility means at least 40% or more, preferably 60% or more, more preferably 80% or more, more preferably 90% or more, still more preferably 95% or more, and particularly preferably 97% or more. This refers to the sequence homology (for example, 98 to 99%). The identity of amino acid sequences can be determined by, for example, the algorithm B LAST (Proc. Natl. Ac ad. S ci. USA 8 7: 2264-2268, 1 990, Proc. Na tl. Ac a d. S c in USA 90: 587 3-58 77, 1 9 3 3). Based on this algorithm, a program called B LAS TX has been developed (Alt s c hul eta 1. J. Mo 1. B i o l. 2 1 5: 40 3— 4 10, 1 990). When analyzing the amino acid sequence by B LAS TX, the parameter 1 is, for example, s corre = 50 and wo rd lenght = 3. When using BLAST and Gappe ED BLAST programs, use the default parameters of each program. Specific methods of these analysis methods are publicly known, http: ZZwww.ncbi.n1m.nih.gov.).
L I V 1に類似するタンパク質の調製は、 別の公知手段によることも可能であ る。 例えば、 ェキソヌクレア一ゼを用いる d e l e t i o n mu t a n t作製 法、 カセッ ト変異法等の s i t e— d i r e c t e d mu t a g e n e s i s によって L I V I DNAを人為的に改変させ、 該改変 L I V 1 DNAを用い て、 所望タンパク質を調製することができる。  Preparation of a protein similar to L I V 1 can also be performed by other known means. For example, it is possible to artificially modify LIVI DNA by site-directed mu tagenesis such as deletion mu tant production method using exonuclease and cassette mutation method, and to prepare desired protein using the modified LIV 1 DNA. it can.
「亜鉛イオン要求性タンパク質の発現及び Z又は機能を調節する物質」 として 利用可能な物質の別の好適な例として、 配列番号 1、 3又は 5記載の配列からな る DNAを挙げることができる。 上記 DNAはタンパク質 L I V 1をコー 'ドして おり、 細胞内に導入されれば L I V 1タンパク質発現を通じて間接的に亜鉛ィォ ン要求性タンパク質、 例えば S n a i 1の活性を調製し得る物質である。 Another preferred example of a substance that can be used as a substance that regulates the expression and Z or function of a zinc ion-requiring protein is the sequence described in SEQ ID NO: 1, 3, or 5. DNA. The above DNA encodes the protein LIV 1, and if introduced into a cell, it is a substance that can indirectly adjust the activity of a zinc-requiring protein, such as Snai 1, through LIV 1 protein expression. .
上記 DNAは、 配列番号 1、 3又は 5記載の配列の一部をプローブとし、 当業 者に周知のハイブリダィゼーシヨン技術によって、 L I V 1が発現している細胞 の cDN Aから調製することができる。 また、 配列番号 1、 3又は 5記載の配列 の一部をプライマーとし、 mRNAから RT— P CRを実施して得ることもでき る。 あるいは市販の DNA合成機を用いて人工的に合成してもよい。  The above DNA should be prepared from cDNA of cells expressing LIV 1 by a hybridization technique well known to those skilled in the art using a part of the sequence shown in SEQ ID NO: 1, 3 or 5 as a probe. Can do. It can also be obtained by performing RT-PCR from mRNA using a part of the sequence shown in SEQ ID NO: 1, 3 or 5 as a primer. Alternatively, it may be artificially synthesized using a commercially available DNA synthesizer.
さらに、上記 DNAに類似する DNAもまた、 「亜鉛イオン要求性タンパク質の 発現及びノ又は機能を調節する物質」 の一例である。 該類似する DNAとして、 配列番号 1、 3又は 5記載の配列とス トリンジヱントな条件でハイブリダイズす る DN Aであって、 亜鉛イオン要求性タンパク質の発現及び/又は機能を調節す る物質、 例えば S n a i 1の活性を調節し得るタンパク質をコードした DNAを 挙げることができる。 この DN Aの調製法は既に上述したとおりである。  Furthermore, DNA similar to the above DNA is also an example of “a substance that regulates the expression and function of a zinc ion-requiring protein”. The similar DNA is a DNA that hybridizes under stringent conditions with the sequence shown in SEQ ID NO: 1, 3, or 5, and is a substance that regulates the expression and / or function of a zinc ion-requiring protein, for example, A DNA encoding a protein capable of regulating the activity of Snai 1 can be mentioned. The method for preparing this DNA is as described above.
上述した各 DNAを使用するときは、 適当なベクターに挿入して用いることが できる。 ベクターに揷入したものも本発明の態様の一つである。 使用するべクタ 一は、 目的に応じて適当なベクターを選択することができる。 具体的には、 哺乳 動物由来のベクター (例えば、 p c DNA 3 (インビトロジヱン社製) や p EG F— BOS (Nu c l e i c Ac i d s . R e s ., 1 8 ( 1 7), p. 5 32 2, 1 990)、 p EF、 p CDM8、 p CXN)、 昆虫細胞由来のベクター (例 は 「B a c— t-o— BAC b a c u l o v i r u s e x p r e s s i o n s y s t e m」 (インビトロジェン社製)、 p B a c PAK8)、植物由来の発現べ クタ一 (例えば ρΜΗ 1、 pMH2)、 動物ウィルス由来のベクター (例えば、 p HSV、 pMV、 pAd e x L c w)、 レトロウイノレス由来のベクター (例えば、 p Z I P n e o )、 酵母由来のベクター (例えば「P i c h i a E x p r e s s i o n K i t J (インビトロジェン社製)、 pNV l l、 S P— Q0 1)、 枯草菌 由来のベクター (例えば p P L 608、 p KTH 50)、 大腸菌べクタ一 (M 1 3 系べクター、 p UC系ベクター、 p BR 3 22、 p B l u e s c r i p t^ p C R-S c r i p t) 等を挙げることができる。 本発明において、 哺乳動物細胞内 で発現可能なベクタ一を用いることが好ましく、 又、 発現ベクターを用いること が好ましい。 ベクターを細胞へ導入するには、 例えば、 リン酸カルシウム法 (V i r o l o g y, Vo l . 5 2, p. 456, 1 973)、 DEAEデキス トラン 法、 カチォニックリボソーム DOTAP (ロッシュ . ダイァグノスティックス社 製)を用いた方法、エレク ト口ポレーシヨン法(N u c 1 e i c Ac i d s R e s . , Vo l . 1 5, p. 1 3 1 1, 1 98 7)、 リポフエクション法 (J. C 1 i n. B i o c h em. N u t r . , Vo l . 7, p. 1 75, 1 98 9)、 ゥ ィルスによる感染導入方法 (pMX、 pMSCV等; S c i . Am. , p. 34, 1 994)、 パーティクルガン等から選択することにより行うことができる。When using each DNA described above, it can be used by inserting it into an appropriate vector. A product inserted into a vector is also one embodiment of the present invention. The vector to be used can select an appropriate vector according to the purpose. Specifically, vectors derived from mammals (for example, pc DNA 3 (manufactured by Invitrogene) and pEG F- BOS (Nucleic Acids. Res., 1 8 (17), p. 5 32 2, 1 990), p EF, p CDM8, p CXN), insect cell-derived vectors (eg “B ac-to- BAC baculovirus expression system” (Invitrogen), p B ac PAK8), plant-derived expression vectors 1 (for example, ρΜΗ1, pMH2), a vector derived from an animal virus (for example, p HSV, pMV, pAd ex L cw), a vector derived from retrowinoles (for example, p ZIP neo), a vector derived from yeast (for example, “P ichia E xpression Kit J (manufactured by Invitrogen), pNV ll, SP—Q0 1), vectors derived from Bacillus subtilis (eg, p PL 608, p KTH 50), E. coli vector (M 1 3 Vector vectors, pUC vectors, pBR322, pBluescript ^ pCRScript) and the like. In the present invention, it is preferable to use one vector that can be expressed in mammalian cells, and it is also preferable to use an expression vector. In order to introduce a vector into cells, for example, calcium phosphate method (Virology, Vol. 52, p. 456, 1 973), DEAE dextran method, Cationic ribosome DOTAP (Roche. Diagnostics) , Electo mouth position method (Nuc 1 eic Acids Res., Vo l. 1 5, p. 1 3 1 1, 1 98 7), lipofection method (J. C 1 B ioch em. Nutr., Vo l. 7, p. 1 75, 1 98 9), virus introduction method (pMX, pMSCV, etc .; S ci. Am., p. 34, 1 994) ), By selecting from a particle gun or the like.
「亜鉛イオン要求性タンパク質の発現及び Z又は機能を調節する物質」、例えば S n a i 1の活性を抑制する剤の一例として、 L I V 1をコードする DNA又は mRNAを標的とするアンチセンスオリゴヌクレオチドを挙げることができる。 L I V 1をコードする DNAに対するアンチセンスオリゴヌクレオチドは、 内在 する L I V 1遺伝子の発現を妨げ、 S n a i 1活性を負に制御すると考えられる。 このようなアンチセンスオリゴヌクレオチドとして、 配列番号 1、 3又は 5記載 の配列からなる D N A又は該 D N Aから生成される m R N Aを標的配列とするァ ンチセンスオリゴヌクレオチドを挙げることができる。 一例としては、 配列番号 7又は 8に記載するオリゴヌクレオチドを挙げることができる。 これ以外にも、 上記配列番号 1、 3又は 5に記載の配列からなる DN A等のいずれかの箇所にハ イブリダィズし L I V 1の発現を有効に阻害できれば、 本発明のアンチセンスォ リゴヌクレオチドに含まれ、 上記配列番号 1、 3又は 5に記載の配列からなる D N A又は対応する mRNAと完全に相補的でなくてもよい。 “Substances that regulate the expression and Z or function of a zinc ion-requiring protein”, for example, an antisense oligonucleotide that targets DNA or mRNA encoding LIV 1 as an example of an agent that suppresses the activity of Snai 1 be able to. Antisense oligonucleotides to DNA encoding L I V 1 are thought to prevent expression of the endogenous L I V 1 gene and negatively regulate Snai 1 activity. Examples of such antisense oligonucleotides include antisense oligonucleotides having a DNA sequence consisting of the sequence set forth in SEQ ID NO: 1, 3 or 5 or an mRNA sequence generated from the DNA sequence as a target sequence. As an example, the oligonucleotide described in SEQ ID NO: 7 or 8 can be mentioned. In addition to this, it is included in the antisense oligonucleotide of the present invention as long as it hybridizes to any location such as DNA consisting of the sequence set forth in SEQ ID NO: 1, 3 or 5 and can effectively inhibit LIV 1 expression. It does not have to be completely complementary to DNA consisting of the sequence set forth in SEQ ID NO: 1, 3 or 5 or the corresponding mRNA.
上記アンチセンスオリゴヌクレオチドは、 目的に応じ、 適当なベクターに挿入 して用いることができる。 例えば遺伝子治療に応用する目的であれば、 レトロゥ イノレスベクター、 アデノウイルスベクタ一、 ワクシニアゥイノレスべクタ一等のゥ ィルスベクターや、 カチォニックリボソーム、 リガンド DNA複合体等の非ウイ ルスベクター等の中から、 適宜選択可能である。 また、 キャリアーを用いずに、 裸のプラスミ ド DNA (n a k e d p DNA) として大容量の水溶液とともに 投与する方法をとることも考えられる。 The antisense oligonucleotide can be used by inserting it into an appropriate vector depending on the purpose. For example, for the purpose of gene therapy, retrovirus vector, adenovirus vector, vaccinia virus vector, etc. It can be appropriately selected from viral vectors, non-viral vectors such as cationic ribosomes and ligand DNA complexes. It is also conceivable to administer a large volume of aqueous solution as naked plasmid DNA without using a carrier.
別の「亜鉛イオン要求性タンパク質の発現及びノ又は機能を調節する物質」、特 に S n a i 1の活性を抑制する剤の例として、 L I V 1をコードする DNAの一 部と同一又 類似する配列を有する二本鎖 RN Aを挙げることができる。 標的遺 伝子配列と同一又は類似した配列を有する二本鎖 RN Aは、 標的遺伝子の発現を 妨げる RNA干渉 (RNA i n t e r f e r e n c e ; RNA i ) を引き起こ し得る。 RNA iは、 二本鎖 RNA (d s RNA) を細胞内に導入した際に、 そ の RNA配列に対応する細胞内の mRNAが特異的に分解され、 タンパク質とし て発現されなくなる現象をいう。 二本鎖を形成する領域は、 すべての領域におい て二本鎖を形成していてもよいし、一部の領域(例えば両末端又は片方の末端等) がー本鎖等になっていてもよい。 従って、 本発明の二本鎖 RNAにおいても、 二 本鎖でない領域が含まれていてよい。 RNA iに用いられるオリゴ RNAは 10 〜: L 00 b pの RNAが用いられることが多く、 通常 1 9〜23 b pの RNAが 用いられる。 RNA i法は、 Na t u r e , Vo l . 39 1, p. 806, 1 9 98、 P r o c . N a t l . Ac a d. S c i . USA Vo l . 95, p. 1 5502, 1 998、 N a t u r e, Vo l . 395, p. 854, 1 998、 P r o c. N a t l . A c a d. S c i . USA Vo l . 96, p. 5049, 1 99 9、 C e l l , V o l . 9 5, p. 1 0 1 7, 1 998、 P r o c. N a t 1. Ac a d. S c i . USA Vo l . 96, p. 1 45 1 , 1 9 99、 P r o c . N a t l . Ac a d. S c i . U SA V o l . 95, p. 1 3 9 59, 1 998、 N a t u r e C e l l B i o l ., Vo l . 2, p. 70, 200 0等の記載に従って行うことができる。  Another “substance that regulates the expression and function of zinc ion-requiring protein”, particularly as an example of an agent that suppresses the activity of Snai 1, a sequence identical or similar to a portion of DNA encoding LIV 1 And a double-stranded RNA having Double-stranded RNA having a sequence that is the same as or similar to the target gene sequence can cause RNA interference (RNAi) that prevents the expression of the target gene. RNA i refers to a phenomenon in which when a double-stranded RNA (dsRNA) is introduced into a cell, the mRNA in the cell corresponding to the RNA sequence is specifically degraded and is not expressed as a protein. The region that forms a double strand may form a double strand in all regions, or a part of the region (for example, both ends or one end) may be a double strand or the like. Good. Therefore, the double-stranded RNA of the present invention may contain a region that is not double-stranded. Oligo RNA used for RNA i is often 10 to: L 00 bp RNA, and usually 19 to 23 bp RNA. RNAi method is Nature, Vo l. 39 1, p. 806, 1 9 98, Proc. N atl. Ac ad. S ci. USA Vo l. 95, p. 1 5502, 1 998, N ature, Vol. 395, p. 854, 1 998, Proc. Natl. A cad. S ci. USA Vol. 96, p. 5049, 1 99 9, Cell, Vol. , p. 1 0 1 7, 1 998, Proc. N at 1. Ac a d. S ci. USA Vo l. 96, p. 1 45 1, 1 9 99, Proc. N atl. Ac a d. Sci. U SA Vol. 95, p. 1 3 9 59, 1 998, Nature Cell Biol., Vol. 2, p. 70, 2000, etc.
「亜鉛イオントランスポーターの発現及び 又は機能を調節する物質」 として は、 例えば該物質が L I V 1の場合、 上記した 「亜鉛イオン要求性タンパク質の 発現及び Z又は機能を調節する物質」 と同様なものが例示される。 As a “substance that regulates the expression and / or function of a zinc ion transporter”, for example, when the substance is LIV 1, Examples are the same as “substances that regulate expression and Z or function”.
「亜鉛イオントランスポーター」 が L I V 1以外の場合、 例えば、 h Z n t— 1、 h Z n t— 2、 h Z n t— 5、 h Z n t— 6、 h Z n t— 7、 h Z I P l、 h Z I P 2、 h Z I P 3、 BAA 9 2 1 00, BAC 04504、 h L i v— 1、 hKE 4、 K I AA 1 265、 K I AA 0062、 h Z i p 4の場合には、 該亜 鉛イオントランスポーターの既知のアミノ酸配列あるいは塩基配列をもとにして、 L I V 1の場合と同様にして各種の 「亜鉛イオントランスポーターの発現及び// 又は機能を調節する物質」 あるいは 「亜鉛イオン要求性タンパク質の発現及び Z 又は機能を調節する物質」 を調製することができる。 - 各種アミノ酸配列及び塩基配列の情報は、 NCB I等で閲覧可能な種々のデー タベースから入手することができる。  When the “Zinc ion transporter” is other than LIV 1, for example, h Z nt— 1, h Z nt— 2, h Z nt— 5, h Z nt— 6, h Z nt— 7, h ZIP l, h ZIP 2, h ZIP 3, BAA 9 2 1 00, BAC 04504, h L iv— 1, hKE 4, KI AA 1 265, KI AA 0062, h Z ip 4 Based on the known amino acid sequence or base sequence, various “substances that regulate the expression and / or function of zinc ion transporter” or “expression and expression of zinc ion-requiring protein” as in LIV 1. A substance that modulates Z or function can be prepared. -Information on various amino acid sequences and nucleotide sequences can be obtained from various databases that can be browsed by NCB I.
本発明の抗原提示細胞の機能制御剤並びに免疫システムが関与する疾患の予 防 '治療薬は、 上記した一連の有効成分 (亜鉛イオン、 亜鉛ィオノフォア、 亜鉛 イオンキレ一ター、 亜鉛イオン要求性タンパク質の発現及び Z又は機能を調節す る物質、 亜鉛イオントランスポーターの発現及びノ又は機能を調節する物質) に 加え、 所望により薬学的に許容される賦形剤、 添加剤を含む。 薬学的に許容ざれ る賦形剤、 添加剤としては、 担体、 結合剤、 香料、 緩衝剤、 増粘剤、 着色剤、 安 定剤、 乳化剤、 分散剤、 懸濁化剤、 防腐剤等が挙げられる。 又、 有効成分として 異なる種類の 「亜鉛イオン濃度を制御する物質」 を併用することもできる。  The agent for controlling the function of antigen-presenting cells and the prevention of diseases involving the immune system according to the present invention includes the above-described series of active ingredients (zinc ion, zinc ionophore, zinc ion killer, expression of zinc ion-requiring protein) And substances that regulate Z or function, substances that regulate the expression and function of zinc ion transporter), and optionally include pharmaceutically acceptable excipients and additives. Pharmaceutically unacceptable excipients and additives include carriers, binders, fragrances, buffers, thickeners, colorants, stabilizers, emulsifiers, dispersants, suspending agents, preservatives, etc. Can be mentioned. Also, different types of “substances that control zinc ion concentration” can be used in combination as active ingredients.
薬学的に許容される担体としては、 例えば、 炭酸マグネシウム、 ステアリン酸 マグネシウム、 タノレク、 砂糖、 ラタ トース、 ぺクチン、 デキス トリン、 澱粉、 ゼ ラチン、 トラガント、 メチルセルロース、 ナトリ ウムカルボキシメチルセルロー ス、 低融点ワックス、 カカオバター等が挙げられる。  Examples of pharmaceutically acceptable carriers include magnesium carbonate, magnesium stearate, tanolec, sugar, ratatoose, pectin, dextrin, starch, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose, and low melting point. Examples include wax and cocoa butter.
さらに、 錠剤は必要に応じて通常の剤皮を施した錠剤、 例えば糖衣錠、 腸溶性 コーティング錠、 フィルムコーティング錠あるいは二層錠、 多層錠とすることが できる。 散剤は、 薬学的に許容される散剤の基剤と共に製剤化される。 基剤とし ては、 タルク、 ラク トース、 澱粉等が挙げられる。 ドロップは水性又は非水性の 基剤と一種またはそれ以上の薬学的に許容される拡散剤、 懸濁化剤、 溶解剤等と 共に製剤化できる。 カプセルは、 有効成分となる化合物を薬学的に許容される担 体と共に中に充填することにより製造できる。 当該化合物は薬学的に許容される 賦形剤と共に混合し、または賦形剤なしで力プセルの中に充填することができる。 カシエ剤も同様の方法で製造できる。 本発明を座剤として調製する場合、 植物油 (ひまし油、 ォリーブ油、 ピーナッツ油等) や鉱物油 (ワセリン、 白色ワセリン 等)、 ロウ類、 '部分合成もしくは全合成グリセリン脂肪酸エステル等の基剤と共に 通常用いられる手法によって製剤化される。 Furthermore, the tablets can be made into tablets with ordinary coatings as necessary, for example, sugar-coated tablets, enteric-coated tablets, film-coated tablets, double-layer tablets, and multilayer tablets. Powders are formulated with a pharmaceutically acceptable powder base. Examples of bases include talc, lactose, and starch. Drops are aqueous or non-aqueous It can be formulated together with a base and one or more pharmaceutically acceptable diffusing agents, suspending agents, solubilizing agents and the like. Capsules can be produced by filling a compound as an active ingredient together with a pharmaceutically acceptable carrier. The compounds can be mixed with pharmaceutically acceptable excipients or filled into force capsules without excipients. A cachet agent can be produced by a similar method. When the present invention is prepared as a suppository, it is usually used together with a base such as vegetable oil (castor oil, olive oil, peanut oil, etc.), mineral oil (petrol, white petrolatum, etc.), waxes, 'partially synthesized or fully synthetic glycerin fatty acid ester. Formulated according to the technique used.
注射用液剤としては、 溶液、 懸濁液、 乳剤等が挙げられる。 例えば、 水溶液、' 水一プロピレングリコー^^溶液等が挙げられる。 液剤は、 水を含んでも良い、 ポ リエチレンダリコール及びノまたはプロピレンダリコールの溶液の形で製造する こともできる。  Examples of injection solutions include solutions, suspensions, and emulsions. For example, an aqueous solution, a water-propylene glycol solution, etc. are mentioned. The solution can also be produced in the form of a solution of polyethylene and / or propylene dalicol, which may contain water.
経口投与に適切な液剤は、 有効成分となる化合物を水に加え、 着色剤、 香料、 安定化剤、甘味剤、溶解剤、増粘剤等を必要に応じて加え製造することができる。 また経口投与に適切な液剤は、 当該化合物を分散剤とともに水に加え、 粘重にす ることによつても製造できる。 増粘剤としては、 例えば、 薬学的に許容される天 然または合成ガム、 レジン、 メチルセルロース、 ナトリ ウムカルボキシメチルセ ルロースまたは公知の懸濁化剤等が挙げられる。  A solution suitable for oral administration can be produced by adding a compound as an active ingredient to water and adding a colorant, a fragrance, a stabilizer, a sweetener, a solubilizer, a thickener and the like as necessary. A solution suitable for oral administration can also be produced by adding the compound together with a dispersant to water to make it viscous. Examples of the thickener include pharmaceutically acceptable natural or synthetic gum, resin, methylcellulose, sodium carboxymethylcellulose, or a known suspending agent.
局所投与剤としては、 上記の液剤及び、 クリーム、 エアロゾル、 スプレー、 粉 剤、 ローション、 軟膏等が挙げられる。 上記の局所投与剤は、 有効成分となる化 合物と薬学的に許容される希釈剤及び担体と混合することによって製造できる。 軟膏及びクリームは、 例えば、 水性または油性の基剤に増粘剤及び Zまたはゲル 化剤を加えて製剤化する。 該基剤としては、 例えば、 水、 液体パラフィン、 植物 油等が挙げられる。 増粘剤としては、 例えばソフ トパラフィン、 ステアリン酸ァ ノレミニゥム、 セ トステアリノレアノレコール、 プロピレングリ コーノレ、 ポリエチレン グリコール、 ラノリ ン、 水素添加ラノリン、 蜜蠟等が挙げられる。 局所投与剤に は、 必要に応じて、 ヒ ドロキシ安息香酸メチル、 ヒ ドロキシ安息香酸プロピル、 クロ口クレゾール、 ベンザルコニゥムクロリ ド等の防腐剤、 細菌増殖防止剤を添 加することもできる。 ローションは、 水性又は油性の基剤に、 一種類またはそれ 以上の薬学的に許容される安定剤、懸濁化剤、乳化剤、拡散剤、増粘剤、着色剤、 香料等を加えることができる。 Examples of the topical administration agent include the above-mentioned liquids, creams, aerosols, sprays, powders, lotions, ointments and the like. The above-mentioned topical preparation can be produced by mixing a compound as an active ingredient with a pharmaceutically acceptable diluent and carrier. Ointments and creams are formulated, for example, by adding a thickener and a Z or gelling agent to an aqueous or oily base. Examples of the base include water, liquid paraffin, and vegetable oil. Examples of the thickening agent include soft paraffin, anolenomium stearate, cetostellenoreanolol, propylene glycolate, polyethylene glycol, lanolin, hydrogenated lanolin, and beeswax. For topical administration, if necessary, methyl hydroxybenzoate, propyl hydroxybenzoate, Preservatives such as black mouth cresol and benzalkonium chloride and bacterial growth inhibitors can be added. Lotions can contain one or more pharmaceutically acceptable stabilizers, suspending agents, emulsifiers, diffusing agents, thickeners, colorants, flavors, etc., in an aqueous or oily base. .
かく して得られる本発明の抗原提示細胞の機能制御剤並びに免疫システムが関 与する疾患の予防 ·治療薬は、 経口または非経口的に投与される。  The antigen-presenting cell function-controlling agent of the present invention and the prophylactic / therapeutic agent for diseases related to the immune system are administered orally or parenterally.
経口的に投与する場合、 通常当分野で用いられる投与形態で投与することがで きる。 非経口的に投与する場合には、 局所投'与剤 (経皮剤等)、 直腸投与剤、 注射 剤、 経鼻剤等の投与形態で投与することができる。  When administered orally, it can be administered in a dosage form commonly used in the art. When administered parenterally, it can be administered in a dosage form such as a topical administration agent (transdermal agent, etc.), a rectal agent, an injection, or a nasal agent.
経口剤または直腸投与剤としては、 例えばカプセル、 錠剤、 ピル、 散剤、 ドロ ップ、 カシエ剤、 座剤、 液剤等が挙げられる。 注射剤としては、 例えば、 無菌の 溶液又は懸濁液等が挙げられる。局所投与剤としては、例えば、クリーム、軟膏、 ローション、 経皮剤 (通常のパッチ剤、 マ トリクス剤) 等が挙げられる。  Examples of the oral preparation or rectal preparation include capsules, tablets, pills, powders, drops, cachets, suppositories, and liquids. Examples of injections include sterile solutions or suspensions. Examples of topical agents include creams, ointments, lotions, transdermal agents (ordinary patches, matrix agents) and the like.
投与量、 投与回数は使用する亜鉛イオン濃度を制御し得る物質の種類、 患者の 症状、 年齢、 体重、 投与形態等によって異なり適宜設定され得る。  The dose and the number of doses vary depending on the type of substance that can control the zinc ion concentration to be used, the patient's symptoms, age, weight, dosage form, etc., and can be appropriately set.
「亜鉛イオン」、 「亜鉛イオンキレーター」、 「亜鉛イオン要求性タンパク質の発 現及び/又は機能を調節する物質」 及び 「亜鉛イオントランスポーターの発現及 び Z又は機能を調節する物質」 はいずれも、 ヒ トをはじめゥシ、 ゥマ、 ィヌ、 マ ウス、 ラット等の哺乳動物において、 抗原提示細胞の機能、 具体的には抗原提示 に関係する分子の発現、 L P S等による抗原提示細胞の活性化、 サイト力イン産 生、 抗原の取り込み、 遊走能等を制御することが可能であり、 従って、 抗原提示 細胞の機能に関与する疾患、 具体的には免疫システムが関与する疾患を予防 ·治 療することが可能となる。免疫システムが関与する疾患としては、自己免疫疾患、 アレルギー疾患、 癌、 臓器移植の際の拒絶反応、 感染や腫瘍形成による免疫応答 の低下、 ワクチン使用時の異常な免疫応答 (例えば副作用) 等が挙げられる。 例 えば本発明は、 S I R S (全身性炎症反応症候群)、 全身性エリテマトーデス、 混 合型結合組織病、 慢性関節リウマチ、 シエーダレン症候群、 リウマチ熱、 グッド パスチヤ一症候群、 バセドウ病、 橋本病、 アジソン病、 自己免疫性溶血性貧血、 特発性血小板减少性紫斑病、 重症筋無力症、 潰瘍性大腸炎、 クローン病、 交換性 眼炎、 多発性硬化症、 乾癬、 アナフィラキシーショ ック、 アレルギー性鼻炎、 ァ レルギ一性結膜炎、 気管支喘息、 奪麻疹、 ア トピー性皮膚炎等を処置するため、 臓器移植時の拒絶反応の抑制のため移植前処理、 移植後投与のため、 さらには癌 に対する免疫応答制御等のために使用できると期待される。 癌としては、 乳癌、 前立腺癌、 膝'癌、 胃癌、 肺癌、 大腸癌 (結腸癌、 直腸癌、 肛門癌)、 食道癌、 十二 指腸癌、 頭頸部癌 (舌癌、 咽頭癌)、 脳腫瘍、 神経鞘腫、 非小細胞肺癌、 肺小細胞 癌、 肝臓癌、 腎臓癌、 胆管癌、 子宮癌 (子宮体癌、 子宮頸癌)、 卵巣癌、 膀胱癌'、 皮膚癌、 血管腫、 悪性リンパ腫、 悪性黒色腫、 甲状腺癌、 骨腫瘍、 血管腫、 血管 線維腫、 網膜肉腫、 陰茎癌、 小児固形癌、 力ポジ肉腫、 A I D Sに起因するカポ ジ肉腫、 上顎洞腫瘍、 線維性組織球腫、 平滑筋肉腫、 横紋筋肉腫の癌並びに白血 病等の悪性腫瘍等がそれぞれ挙げられる。 "Zinc ions", "Zinc ion chelators", "Substances that regulate the expression and / or function of zinc ion-requiring proteins" and "Substances that regulate the expression and Z or function of zinc ion transporters" are all In mammals such as humans, horses, horses, dogs, mice, rats, etc., the function of antigen-presenting cells, specifically the expression of molecules related to antigen presentation, the expression of antigen-presenting cells by LPS, etc. It is possible to control activation, site force-in production, antigen uptake, migration ability, etc., and therefore prevent diseases related to the function of antigen-presenting cells, specifically diseases related to the immune system It can be treated. Diseases that involve the immune system include autoimmune diseases, allergic diseases, cancer, rejection during organ transplantation, decreased immune response due to infection and tumor formation, and abnormal immune responses (eg, side effects) when using vaccines. Can be mentioned. For example, the present invention includes SIRS (systemic inflammatory response syndrome), systemic lupus erythematosus, mixed connective tissue disease, rheumatoid arthritis, siedalene syndrome, rheumatic fever, Good Pastier I syndrome, Graves' disease, Hashimoto's disease, Addison's disease, autoimmune hemolytic anemia, idiopathic platelet-poor purpura, myasthenia gravis, ulcerative colitis, Crohn's disease, interchangeable ophthalmitis, multiple sclerosis To treat psoriasis, anaphylactic shock, allergic rhinitis, allergic conjunctivitis, bronchial asthma, measles, atopic dermatitis, etc. It is expected that it can be used for post-administration and for controlling immune responses against cancer. Cancers include breast cancer, prostate cancer, knee cancer, stomach cancer, lung cancer, colon cancer (colon cancer, rectal cancer, anal cancer), esophageal cancer, duodenal cancer, head and neck cancer (tongue cancer, pharyngeal cancer), Brain tumor, schwannoma, non-small cell lung cancer, small cell lung cancer, liver cancer, kidney cancer, bile duct cancer, uterine cancer (uterine body cancer, cervical cancer), ovarian cancer, bladder cancer ', skin cancer, hemangioma, Malignant lymphoma, Malignant melanoma, Thyroid cancer, Bone tumor, Hemangioma, Angiofibroma, Retinal sarcoma, Penile cancer, Pediatric solid cancer, Force positive sarcoma, Capodisarcoma due to AIDS, Maxillary sinus tumor, Fibrous histiosphere Tumor, leiomyosarcoma, rhabdomyosarcoma cancer and malignant tumor such as leukemia.
また、 亜鉛ィオノフォア等の薬剤により亜鉛イオンを抗原提示細胞内に導入す ることにより L P S等による抗原提示細胞の活性化を抑制したり、 サイ ト力イン 産生を抑制したりすることが可能となり、 さらに T h 1及び T h 2のバランス、 C T L、 B細胞活性化の制御も可能となる。 すなわち、 本発明の抗原提示細胞の 機能制御剤並びに免疫システムが関与する^患の予防 ·治療薬は、 抗原提示細胞 内への亜鉛イオンの導入により治療効果が認められるような各種疾患に罹患した 患者をも投与対象とすることができる。  In addition, by introducing zinc ions into antigen-presenting cells using a drug such as zinc ionophore, it becomes possible to suppress activation of antigen-presenting cells by LPS or the like, and to suppress site-in production. Furthermore, it is possible to control the balance of T h 1 and T h 2 and the activation of CTL and B cells. In other words, the agent for preventing and treating diseases associated with the function-controlling agent of the antigen-presenting cell and the immune system of the present invention suffers from various diseases whose therapeutic effect is recognized by the introduction of zinc ions into the antigen-presenting cell. Patients can also be the subject of administration.
さらに、 亜鉛イオン濃度を制御することによつて抗原提示細胞の機能を制御す ることができる、 という本発明者らが得た知見により、 細胞内の亜鉛イオン濃度 を測定することによって抗原提示細胞の機能制御が可能な化合物をスクリーニン グすることができる。 例えば以下の工程により本スクリーユング方法は実施され る。  Furthermore, based on the knowledge obtained by the present inventors that the function of antigen-presenting cells can be controlled by controlling the zinc ion concentration, antigen-presenting cells can be measured by measuring the intracellular zinc ion concentration. It is possible to screen for compounds that can control the functions of these. For example, the screening method is implemented by the following steps.
1 ) 抗原提示細胞を 2群に分け一方を試験化合物で処理する工程 (未処理の残る 一方を対照とする) 2 ) 処理後の細胞、 及び未処理の対照細胞についてそれぞれ細胞内の亜鉛 オン 濃度を測定する工程 1) Dividing antigen-presenting cells into two groups and treating one with the test compound (the remaining untreated is the control) 2) A step of measuring the intracellular zinc-on concentration of treated cells and untreated control cells.
3 ) 対照細胞の亜鉛イオン濃度と比較して有意に亜鉛イオン濃度に変動があった ものを選択し抗原提示細胞の機能を制御し得る化合物と認定する工程。  3) A step of selecting a compound whose zinc ion concentration is significantly different from that of the control cell and certifying it as a compound capable of controlling the function of the antigen-presenting cell.
抗原提示細胞としては、上記のものが用いられる。好ましくは樹状細胞である。 適当に 2群に分け一方を試験化合物で処理する。 もう一方は処理せず対照細胞と して使用する。さらに亜鉛イオン濃度に影響を及ぼすことが知られている化合物、 例えば亜鉛イオンキレーターである T P E N等をポジティブな対照化合物として 用いてもよい。 試験化合物は、 公知の化合物であっても今後開発される新規な化 合物であってもよい。 また、 低分子化合物であっても高分子化合物であってもか まわない。 ここで低分子化合物とは分子量 3 0 0 0未満程度の化合物であって、 例えば医薬品として通常使用し得る有機化合物及びその誘導体や無機化合物が挙 げられ、 有機合成法等を駆使して製造される化合物やその誘導体、 天然由来の化 合物やその誘導体、 プロモーター等の小さな核酸分子や各種の金属等であり、 望 ましくは医薬品として使用し得る有機化合物及びその誘導体、 核酸分子をいう。 また、 高分子化合物としては分子量 3 0 0 0以上程度の化合物であって、 タンパ ク質、 ポリ核酸類、 多糖類、 及びこれらを組み合わせたもの等が挙げられ、 望ま しくはタンパク質である。 これらの低分子化合物あるいは高分子化合物は、 公知 のものであれば商業的に入手可能であるか、 各報告文献に従って採取、 製造、 精 製等の工程を経て得ることができる。 これらは、 天然由来であっても、 また遺伝 子工学的に調製されるものであってもよく、 また半合成等によっても得ることが できる。  As the antigen-presenting cells, those described above are used. Dendritic cells are preferred. Appropriately divide into two groups and treat one with the test compound. The other is not treated and used as a control cell. Furthermore, compounds known to affect the zinc ion concentration, such as zinc ion chelator TPEN, may be used as a positive control compound. The test compound may be a known compound or a new compound to be developed in the future. Further, it may be a low molecular compound or a high molecular compound. Here, the low molecular weight compound is a compound having a molecular weight of less than 300, for example, an organic compound that can be normally used as a pharmaceutical, a derivative thereof, and an inorganic compound. Compounds, derivatives thereof, naturally derived compounds, derivatives thereof, small nucleic acid molecules such as promoters and various metals, and preferably organic compounds that can be used as pharmaceuticals, derivatives thereof, and nucleic acid molecules. The high molecular compound is a compound having a molecular weight of about 300 or more, and includes proteins, polynucleic acids, polysaccharides, and combinations thereof, and is preferably a protein. These low-molecular compounds or high-molecular compounds can be obtained commercially if they are known, or can be obtained through steps such as collection, production, and purification according to each report literature. These may be of natural origin, may be prepared by genetic engineering, or may be obtained by semisynthesis or the like.
細胞の試験化合物での処理時間は、 用いる細胞や試験化合物の種類や濃度によ つて適宜設定される。 T P E N等のポジティブな対照化合物を用いる場合にはそ の対照化合物で処理した場合に亜鉛ィオン濃度に変動が確認されることを目安に して行うことができる。 細胞を処理する試験化合物の濃度もまた、 用いる細胞や 試験化合物の種類、 処理時間によって適宜設定される。 細胞内の亜鉛イオン濃度の測定は、 通常当分野で実施されている方法を利用し て、 またそれに準じて行うことができる。 例えば原子吸光法 (フレーム法) によ る直接測定、 特異プローブ (例えば蛍光試薬) を用いた蛍光分光光度計によって 測定することができる。 The treatment time of the cells with the test compound is appropriately set depending on the type of cells used and the test compound. In the case of using a positive control compound such as TPEN, it can be carried out with reference to the fact that fluctuations in the zincion concentration are confirmed when treated with the control compound. The concentration of the test compound for treating the cells is also set appropriately depending on the type of cells used, the type of test compound, and the treatment time. The measurement of intracellular zinc ion concentration can be carried out by using a method usually carried out in this field or according to it. For example, it can be measured directly by an atomic absorption method (frame method) or by a fluorescence spectrophotometer using a specific probe (for example, a fluorescent reagent).
実施例  Example
以下、 実施例にそって本発明をさらに詳細に説明するが、 これら実施例は本発 明の範囲を何ら限定するものではない。 本出願全体を通して引用されたすベての 刊行物は参照として本明細書に組み入れられる。 また、 本発明において使用する 試薬や装置、 材料は特に言及されない限り、 商業的に入手可能である。  Hereinafter, the present invention will be described in more detail with reference to examples, but these examples do not limit the scope of the present invention in any way. All publications cited throughout this application are hereby incorporated by reference. In addition, the reagents, devices, and materials used in the present invention are commercially available unless otherwise specified.
実施例 1 Example 1
抗原提示細胞の一つである樹状細胞を亜鉛イオンキレーターである N, N, N,, N' —テトラキス (2—ピリジルメチル) エチレンジァミン (TPEN) で処理 した場合の、 細胞表面上の MHCクラス I分子、 クラス I I分子及び CD 86分 子の発現状況を調べた。  MHC class on the cell surface when dendritic cells, one of the antigen-presenting cells, are treated with the zinc ion chelator N, N, N, N '—tetrakis (2-pyridylmethyl) ethylenediamine (TPEN) The expression status of I molecule, class II molecule and CD 86 molecule was examined.
C 5 7 B LZ6マウス (3本クレアより入手) より骨髄細胞を得、 1 0%FC S (EQU I TECH-B I O I n c., # S FB— 30— 1 388)、 顆粒球 マクロファージコロニ一刺激因子 (GM— C S F) を加えた R PMI培地 (S I GMA, R 87 58) で 6日間 37°C、 5 % C O 2環境下で細胞培養し、 骨髄由 来の樹状細胞 (BMDC) を分化誘導した。 この BMDCに TPEN (S I GM A, P 44 1 3 ) を 6時間から 1 2時間、 0〜 2 Mの濃度になるように培地に 添加した。 同時に抗原提示細胞の活性化のコントロールの条件としてリ.ポポリサ ッカライド (L P S ; 1 0 O n g /m L ; S I GMA, L 26 37 , E. C o l i O 55 : B 5) を加えた群も作成した。 1 2時間 (若しくは 6時間)後に細胞 表面に発現してきた MHCクラス I、 クラス I I及び CD 86分子を、 FAC S 解析により評価した。 結果を図 1に示す。 図 1中の数値はそれぞれの分子を高発 現している細胞の割合を示す。 亜鉛イオンキレーターで処理することにより L P Sと同様に BMDC上の MHCクラス I I分子の発現が増強した。 この発現増強 は薬剤の投与量依存的 (処理時間 1 2時間)、 処理時間依存的 (処理濃度 . 5 IX M) に見られた。 Bone marrow cells were obtained from C 5 7 B LZ6 mice (obtained from 3 Claires), 10% FC S (EQU I TECH-B IOInc., # S FB—30— 1 388), granulocytes, macrophage colonies Cells were cultured in RPMI medium (SI GMA, R 87 58) with stimulating factor (GM—CSF) for 6 days at 37 ° C in a 5% CO 2 environment, and bone marrow-derived dendritic cells (BMDC) were cultured. Differentiation was induced. To this BMDC, TPEN (SI GM A, P 44 1 3) was added to the medium at a concentration of 0 to 2 M for 6 to 12 hours. At the same time, a group with addition of lipopolysaccharide (LPS; 10 Ong / ml; SI GMA, L 26 37, E. coli O 55: B 5) was also prepared as a condition for controlling the activation of antigen-presenting cells. did. 1 MHC class I, class II and CD86 molecules that had been expressed on the cell surface after 2 hours (or 6 hours) were evaluated by FACS analysis. The results are shown in Figure 1. The numbers in Fig. 1 indicate the percentage of cells that highly express each molecule. Treatment with a zinc ion chelator enhanced the expression of MHC class II molecules on BMDC in the same manner as LPS. This enhanced expression Was observed in a dose-dependent manner (treatment time 12 hours) and treatment time-dependent (treatment concentration .5 IX M).
実施例 2 Example 2
実施例 1と同様にして調製した BMDCに亜鉛ィオノフォアであるピリチオン (5 / M ; Mo l e c u l a r p r o b e s , ρ— 24 1 9 3) と Z n 2+ (Z n S04) を培養液中に最終濃度 5 μΜになるように加え、 さらに L P S ( 1 0 O n gZmL') で刺激した。 6時間後、 細胞表面に発現してきた MH Cクラス I I及び CD 8 6分子を、 F AC S解析により評価した。 結果を図 2に示す。 図 2 中の数値はそれぞれの分子を高発現している細胞の割合を示す。 また同時に BM DCの I L— 1 2産生を 4%パラホルムアルデヒ ドで固定化した後に抗 I L— 1 2 p 4 0饥体 ( B D . B i o s c i e n c e P h a r m i n g e n, # 5 54 4 7 9) とアイソタイプコントロール抗体 (BD. B i o s c i e n c e P h a r m i n g e n, # 5 5 3 9 2 5) を用いて細胞内染色法により検出した。 図 中の数値は I L— 1 2を産生している細胞群の割合を示す。 BMDC prepared in the same manner as in Example 1 was added with zincthiophore pyrithione (5 / M; Mollecularprobes, ρ—24 1 93) and Zn 2+ (Z n S0 4 ) in the final concentration of 5 In addition, it was stimulated with LPS (10 OngZmL '). Six hours later, MHC class II and CD86 molecules expressed on the cell surface were evaluated by FACS analysis. The result is shown in figure 2. The numbers in Figure 2 indicate the percentage of cells that highly express each molecule. At the same time, IL-12 production of BM DC was immobilized with 4% paraformaldehyde, followed by anti-IL-12 p40 rod (BD. Bioscience P harmingen, # 5 54 4 7 9) and isotype control antibody. (BD. Bioscience P harmingen, # 5 5 3 9 2 5) was used for detection by intracellular staining. The numbers in the figure show the percentage of cells producing IL-12.
L P S刺激による BMDCの活性化に伴う MHC I I、 CD 8 6分子の細胞表 面での発現増強や I L— 1 2のサイトカイン産生が亜鉛ィオノフォアの存在下で は全く見られなかった。  The expression of MHC I I and CD 86 molecules on the cell surface and the production of IL-12 cytokines following activation of BMDC by LPS stimulation were not observed in the presence of zinc ionophore.
実施例 1及び 2と同様な実験 ( i n V i t r o系) において 1〜 1 0 μΜの 亜鉛イオン若しくは T P EN処理で樹状細胞の活性化若しくは不活性化が見られ た。  In the same experiment as in Examples 1 and 2 (in Vitro system), activation or inactivation of dendritic cells was observed by treatment with 1 to 10 μΜ of zinc ions or T P EN.
実施例 3 Example 3
亜鉛イオン濃度を制御する物質としてマウスの L i V 1を発現するレトロウイ ルスベクターを用いて L P S刺激による MHCクラス I I分子の発現増強に及ぼ す影響を調べた。  Using a retroviral vector that expresses mouse Li V 1 as a substance that controls zinc ion concentration, the effect of LPS stimulation on the enhancement of MHC class I I molecule expression was examined.
理研より提供されたマウス L i V 1遺伝子 (mL i v 1、 遺伝子配列:配列番 号 5、 アミノ酸配列:配列番号 6) を含むクローン (m f j 046 2 3) から制 限酵素 X h o I及び N o t Iにより mL i V 1遺伝子を切断回収し、 東大医科研 の北村俊雄教授が作成し、 提供された p MX— I RE S— GF Pレトロゥ 'ィルス ベクターに組み込んだ。 この pMX— I R E S-GF P-mL i v 1ベクターを 米国スタンフォード大学の G a r r y P. N o 1 a n博士より提供された P h o e n i x細胞にリポフエクトァミン ( I n v i t r o g e n) を用いて導入 しウィルスを発現させた。 24時間から 48時間後に、 このウィルスを含む培養 上澄みを回収し、 GM— CS F 〔GM— C S F産生細胞株 (GM—C S F/CH O) を培養し'、 その培養上清を 0. 3%になるように培地に加える〕、 FC S (1 0%)、 p r o t am i n e s u 1 f a t e (S I GMA; 2 μ g /m L) を添 カロして、 ウィルス感染液とした。 C 5 7 B LZ6マウス (S本クレア) 骨髄より 調製した BMDCに D a y 2及び D a y 4に培地をウィルス感染液に交換し B M DCに感染させた。 D a y 6でウィルスフリーの培地に交換し、さらに L P S (S I GMA; 100 n g/mL) で刺激した。 刺激後 6時間で細胞を回収し、 細胞 表面の MHCクラス I I分子の発現レベルを F AC Sにより調べた。 ここで、 L P S刺激しない細胞群をコントロールとして、 また G F P陽性細胞を mL i V 1 が強制発現している細胞として扱った。 Restriction enzymes X ho I and Not from clones (mfj 046 2 3) containing the mouse Li V 1 gene (mL iv 1, gene sequence: SEQ ID NO: 5, amino acid sequence: SEQ ID NO: 6) provided by RIKEN The mL i V 1 gene was cleaved and collected by I, and the University of Tokyo Medical Research Institute Produced by Prof. Toshio Kitamura and incorporated into the provided p MX—I RE S—GF P retrovirus vector. This pMX—IRE S-GF P-mL iv 1 vector was introduced into Phoenix cells provided by Dr. Garry P. No 1 an at Stanford University, USA using lipofectamine (Invitrogen) to transfer the virus. Expressed. After 24 to 48 hours, the culture supernatant containing this virus was collected, and GM-CSF [GM-CSF producing cell line (GM-CSF / CHO) was cultured, and the culture supernatant was treated with 0.3% FCS (10%) and prot am inesu 1 fate (SI GMA; 2 μg / ml) were added to obtain a virus infection solution. C 5 7 B LZ6 mouse (S-crea) BMDC prepared from bone marrow was infected with BM DC by replacing the medium with virus infection solution in Day 2 and Day 4. The medium was replaced with virus-free medium with Day 6, and further stimulated with LPS (SI GMA; 100 ng / mL). Cells were collected 6 hours after stimulation, and the expression level of MHC class II molecules on the cell surface was examined by FACS. Here, a cell group not stimulated with LPS was treated as a control, and a GFP positive cell was treated as a cell in which mL i V 1 was forcibly expressed.
L P S刺激による MHCクラス I I分子の発現増強が m L i v 1強制発現細胞 では抑制されることがわかった。  It was found that the enhanced expression of MHC class I I molecules by L PS stimulation was suppressed in cells with forced expression of m L i v 1.
(配列表フリーテキス ト)  (Sequence listing free text)
配列番号 7 : アンチセンス配列  Sequence number 7: Antisense sequence | arrangement
配列番号 8 : アンチセンス配列  Sequence number 8: Antisense sequence | arrangement
産業上の利用可能性  Industrial applicability
抗原提示細胞において、 亜鉛イオン濃度を制御したり、 亜鉛イオン要求性のタ ンパク質及び亜鉛イオントランスポーターの発現や機能を調節したりすることで 抗原提示に関係する分子 (例えば MHCクラス I、 クラス I I、 また CD 86分 子等) を発現増強することができる。 さらに亜鉛ィオノフォア等の薬剤により亜 鉛イオンを抗原提示細胞内に導入することでリポポリサッカライド (L P S) 等 による抗原提示細胞の活性化、 サイト力イン産生を抑制することができる。 従つ て、 菌体の構成成分や合成ステロイドといった従来の免疫賦活化分子や免疫抑制 剤等を使用することなしに、 元来体の中に存在している亜鉛の細胞内レベルを調 節することだけで、 例えば樹状細胞等の抗原提示細胞の活性化を介した免疫系を 増強したり抑制したりすることが可能となる。 本出願は、 日本特許出願、 特願 2005— 023200 (出願日 : 200 5年 1月 3 1日) 'を基礎としており、 その内容は全て本明細書に包含される。 In antigen-presenting cells, molecules involved in antigen presentation (eg, MHC class I, class) by controlling the zinc ion concentration or regulating the expression and function of zinc ion-requiring proteins and zinc ion transporters II, CD 86 molecule, etc.) can be enhanced. Furthermore, introduction of zinc ions into antigen-presenting cells with a drug such as zinc ionophore can suppress activation of antigen-presenting cells and production of cyto force in by lipopolysaccharide (LPS). Follow Therefore, it is only necessary to regulate the intracellular level of zinc originally present in the body without using conventional immunostimulatory molecules or immunosuppressive agents such as bacterial components or synthetic steroids. Thus, for example, it is possible to enhance or suppress the immune system through activation of antigen-presenting cells such as dendritic cells. This application is based on a Japanese patent application, Japanese Patent Application No. 2005-0223200 (filing date: Jan. 3, 2005) ', the entire contents of which are included in this specification.

Claims

請求の範囲 , ' The scope of the claims , '
1. 細胞内の亜鉛イオン濃度を制御し得る物質を有効成分として含有する、 抗原 提示細胞の機能制御剤。 1. a function-controlling agent for an antigen-presenting cell, which contains, as an active ingredient, a substance capable of controlling intracellular zinc ion concentration.
2. 亜鉛イオン濃度を制御し得る物質が亜鉛イオンである、 請求項 1記載の機能 制御剤。  2. The function control agent according to claim 1, wherein the substance capable of controlling the zinc ion concentration is zinc ion.
3. 亜鉛イオンが亜鉛ィオノフォアによって細胞内に導入される、 請求項 2記載 の機能制御剤'。  3. The function control agent according to claim 2, wherein the zinc ion is introduced into the cell by a zinc ionophore.
4. 亜鉛イオン濃度を制御し得る物質が亜鉛イオンキレーダーである、 請求項 1 記載の機能制御剤。  4. The function control agent according to claim 1, wherein the substance capable of controlling the zinc ion concentration is a zinc ion killer.
5.亜鉛イオンキレーターが 2, 3—ジメルカプト一 1—プロパンスルホン酸(D MP S)、 N, N, Ν', N' ーテ トラキス (2—ピリジルメチル) エチレンジァ ミン (ΤΡΕΝ)、 エチレンジァミン (EDTA) 及び Ν— (6—メ トキシー 8— キノ リル) 一 ρ— トルエンスルホンアミ ド (T SQ) からなる群より選択される 少なく とも 1種である、 請求項 4記載の機能制御剤。 5. Zinc ion chelator is 2, 3-dimercapto 1-propanesulfonic acid (DMP S), N, N, Ν ', N'-tetrakis (2-pyridylmethyl) ethylene diamine (ΤΡΕΝ), ethylene diamine (EDTA) ) And Ν- (6-Methoxy 8-quinolyl) 1 ρ-Toluenesulfonamide (T SQ). 5. The function control agent according to claim 4.
6. 亜鉛イオン濃度を制御し得る物質が、 亜鉛イオン要求性タンパク質の発現及 び 又は機能を調節する物質である、 請求項 1記載の機能制御剤。 6. The function control agent according to claim 1, wherein the substance capable of controlling the zinc ion concentration is a substance that regulates the expression and / or function of a zinc ion-requiring protein.
7. 亜鉛イオン要求性タンパク質が R a f — 1、 PKC a、 GEF—H 1、 HD AC、 SQSTM1、 ュビキチン一タンパク質リガーゼ E 3、 ュビキチン共役タ ンパク質 E 2、 メタロチォネィン、 フォスファターゼ、 S n a i 1、 TRAF 6、 P a x i 1 1 i n、 Z y x i n及び J a d e— 1からなる群より選択される少な くとも 1種である、 請求項 6記載の機能制御剤。  7. Zinc ion-requiring proteins are Raf-1, PKCa, GEF—H1, HD AC, SQSTM1, ubiquitin monoprotein ligase E3, ubiquitin-conjugated protein E2, metallothinin, phosphatase, Snai1, TRAF 6. The function control agent according to claim 6, wherein the function control agent is at least one selected from the group consisting of P axi 1 1 in, Z yxin and J ade-1.
8. 亜鉛イオン濃度を制御し得る物質が、 亜鉛イオントランスポーターの発現及 び Z又は機能を調節する物質である、 請求項 1記載の機能制御剤。  8. The function control agent according to claim 1, wherein the substance capable of controlling the zinc ion concentration is a substance that regulates expression and Z or function of a zinc ion transporter.
9. 亜鉛イオントランスポーターが L I Vファミ リーを含むヒ ト Z I P類、 ヒ ト CDF類からなる群より選択される少なく とも 1種である、 請求項 8記載の機能 制御剤。  9. The function control agent according to claim 8, wherein the zinc ion transporter is at least one selected from the group consisting of human Z IPs and human CDFs containing an LI V family.
10. 抗原提示細胞が樹状細胞、マクロファージ、 ランゲルハンス細胞、 B細胞、 胸腺上皮細胞、 滑膜細胞、 血管内皮細胞及びケラチノサイ トからなる群より選ば れる少なくとも 1種である、 請求項 1〜9のいずれか 1項に記載の機能制御剤。10. Antigen-presenting cells are dendritic cells, macrophages, Langerhans cells, B cells, The function control agent according to any one of claims 1 to 9, which is at least one selected from the group consisting of thymic epithelial cells, synoviocytes, vascular endothelial cells, and keratinocytes.
1 1. 抗原提示細胞が榭状細胞である、 請求項 1 0記載の機能制御剤。 1 1. The function control agent according to claim 10, wherein the antigen-presenting cell is a rod-shaped cell.
1 2. 請求項 1〜1 1のいずれか 1項に記載の機能制御剤を含む、 免疫システム が関与する疾患の予防 ·治療薬。  1 2. A prophylactic / therapeutic agent for a disease involving the immune system, comprising the function control agent according to any one of claims 1-11.
1 3. 免疫システムが関与する疾患が、 自己免 疾患、 アレルギー性疾患、 臓器 移植の際の拒 反応、 感染や腫瘍形成による免疫応答の低下及びワクチン使用時 の異常な免疫応答からなる群より選ばれる少なくとも 1種である、 請求項 1 2記 載の予防 ·治療薬。 - 1 3. The disease involving the immune system is selected from the group consisting of self-immune disease, allergic disease, rejection at the time of organ transplantation, decreased immune response due to infection and tumor formation, and abnormal immune response at the time of vaccine use The preventive / therapeutic agent according to claim 12, which is at least one kind. -
14. 細胞内の亜鉛イオン濃度を制御し得る物質を有効成分として含有する、 免 疫システムが関与する疾患の予防 ·治療薬。 14. A prophylactic / therapeutic drug for diseases involving an immune system, containing as an active ingredient a substance that can control the intracellular zinc ion concentration.
1 5. 亜鉛イオン濃度を制御し得る物質が亜鉛イオンである、 請求項 14記載の 予防 ·治療薬。  1 5. The prophylactic / therapeutic agent according to claim 14, wherein the substance capable of controlling the zinc ion concentration is zinc ion.
1 6. 亜鉛イオンが亜鉛ィオノフォアによって細胞内に導入されるものである、 請求項 1 5記載の予防 ·治療薬。  1 6. The preventive / therapeutic agent according to claim 15, wherein the zinc ion is introduced into the cell by a zinc ionophore.
1 7. 亜鉛イオン濃度を制御し得る物質が亜鉛イオンキレーターである、 請求項 14記載の予防 ·治療薬。  1 7. The prophylactic / therapeutic agent according to claim 14, wherein the substance capable of controlling the zinc ion concentration is a zinc ion chelator.
1 8. 亜鉛イオンキレーターが 2 , 3—ジメルカプト— 1—プロパンスルホン酸 (DMP S)、 N, N, Ν', N' —テトラキス (2—ピリジルメチル) エチレン ジァミン (T PEN)、 エチレンジァミン (EDTA) 及び N— (6—メ トキシ一 8—キノリル) 一 p—トルエンスルホンアミ ド (TSQ) からなる群より選択さ れる少なくとも 1種である、 請求項 1 7記載の予防 ·治療薬。  1 8. Zinc ion chelator is 2,3-dimercapto-1-propanesulfonic acid (DMP S), N, N, Ν ', N' —tetrakis (2-pyridylmethyl) ethylene diamine (T PEN), ethylene diamine (EDTA) The prophylactic / therapeutic agent according to claim 17, which is at least one selected from the group consisting of N) (6-methoxyl 8-quinolyl) p-toluenesulfonamide (TSQ).
1 9. 亜鉛イオン濃度を制御し得る物質が、 亜鉛イオン要求性タンパク質の発現 及び 又は機能を調節する物質である、 請求項 14記載の予防 ·治療薬。  1 9. The prophylactic / therapeutic agent according to claim 14, wherein the substance capable of controlling the zinc ion concentration is a substance that regulates the expression and / or function of a zinc ion-requiring protein.
20. 亜鉛イオン要求性タンパク質が R a f — 1、 PKC a、 GEF— H l、 H DAC、 SQSTM1、 ュビキチン一タンパク質リガ一ゼ E 3、 ュビキチン共役 タンパク質 E 2、 メタ口チォネイン、 フォスファターゼ、 S n a i 1、 TRAF 6、 P a x i 1 1 i n、 Z y x i n及び J a d e— 1力 らなる群より選択される 少なくとも 1種である、 請求項 1 9記載の予防 ·治療薬。 20. Zinc ion-requiring protein is R af — 1, PKC a, GEF—H l, H DAC, SQSTM1, ubiquitin-protein ligase E 3, ubiquitin-coupled protein E 2, metamouthoneone, phosphatase, S nai 1 , TRAF 10. The prophylactic / therapeutic agent according to claim 19, which is at least one selected from the group consisting of 6, P axi 1 1 in, Z yxin and J ade-1 force.
2 1. 亜鉛イオン濃度を制御し得る物質が、 亜鉛イオントランスポーターの発現 及び Z又は機能を調節する物質である、 請求項 14記載の予防 ·治療薬。  2 1. The prophylactic / therapeutic agent according to claim 14, wherein the substance capable of controlling the zinc ion concentration is a substance that regulates expression and Z or function of a zinc ion transporter.
22. 亜鉛イオントランスポ一ターが L I Vファミ リーを含むヒ ト Z I P類、 ヒ ト CDF類からなる群より選択される少なくとも 1種である、 請求項 2 1記載の 予防 ·治療 。 22. The prophylaxis / treatment according to claim 21, wherein the zinc ion transporter is at least one selected from the group consisting of human Z IPs and human CDFs containing LI family.
23. 免疫システムが関与する疾患が、 自己免疫疾患、 アレルギー性疾患、 臓器 移植の際の拒絶反応、 感染や腫瘍形成による免疫応答の低下及びヮクチン使用時 の異常な免疫応答からなる群より選ばれる少なく とも 1種である、 請求項 1 4〜 22のいずれか 1項に記載の予防 ·治療薬。  23. Diseases involving the immune system are selected from the group consisting of autoimmune diseases, allergic diseases, rejection at the time of organ transplantation, reduced immune response due to infection and tumor formation, and abnormal immune response when using acupuncture The prophylactic / therapeutic agent according to any one of claims 14 to 22, which is at least one kind.
24. 抗原提示細胞が樹状細胞、マクロファージ、 ランゲルハンス細胞、 B細胞、 胸腺上皮細胞、 滑膜細胞、 血管内皮細胞及びケラチノサイ トからなる群より選ば れる少なく とも 1種である、 請求項 14〜23のいずれか 1項に記載の予防 ·治 療薬。  24. The antigen-presenting cell is at least one selected from the group consisting of dendritic cells, macrophages, Langerhans cells, B cells, thymic epithelial cells, synovial cells, vascular endothelial cells, and keratinocytes. The preventive / therapeutic drug according to any one of 1.
25. 抗原提示細胞が樹状細胞である、 請求項 24記載の予防 ·治療薬。  25. The preventive / therapeutic agent according to claim 24, wherein the antigen-presenting cell is a dendritic cell.
26. インビトロで、 細胞内の亜鉛イオン濃度を制御することを含む、 抗原提示 細胞の機能を制御する方法。  26. A method of controlling the function of an antigen-presenting cell, comprising controlling the concentration of zinc ions in a cell in vitro.
27. 亜鉛イオン濃度の制御が亜鉛イオンの細胞内への導入によって行われるも のである、 請求項 26記載の方法。  27. The method according to claim 26, wherein the zinc ion concentration is controlled by introducing zinc ions into cells.
28. 亜鉛イオンの細胞内への導入が亜鉛ィオノフォアによって行われるもので ある、 請求項 27記載の方法。  28. The method according to claim 27, wherein the zinc ion is introduced into the cell by a zinc ionophore.
29. 亜鉛イオン濃度の制御が亜鉛イオンキレーターによって行われるものであ る、 請求項 26記載の方法。  29. The method according to claim 26, wherein the zinc ion concentration is controlled by a zinc ion chelator.
30. 亜鉛イオンキレーターが 2, 3—ジメルカプト _ 1 _プロパンスルホン酸 (DMP S)、 Ν, Ν, Ν', Ν' ーテ トラキス (2—ピリジルメチル) エチレン ジァミン (ΤΡ ΕΝ)、 エチレンジァミン (EDTA) 及び Ν— (6—メ トキシ一 8—キノリル) 一 p— トルエンスルホンアミ ド (T S Q) からなる群より選択さ れる少なくとも 1種である、 請求項 2 9記載の方法。 30. Zinc ion chelator is 2,3-dimercapto _ 1 _propanesulfonic acid (DMP S), Ν, Ν, Ν ', Ν'-Tetrakis (2-pyridylmethyl) ethylene diamine (ΤΡ ΕΝ), ethylene diamine (EDTA) ) And Ν— (6-Methoxy The method according to claim 29, which is at least one member selected from the group consisting of 8-quinolyl) 1-toluenesulfonamide (TSQ).
3 1. 亜鉛イオン濃度の制御が亜鉛イオン要求性タンパク質の発現及ぴ 又は機 能を調節することによって行われるものである、 請求項 26記載の方法。  3. The method according to claim 26, wherein the zinc ion concentration is controlled by regulating the expression or function of a zinc ion-requiring protein.
32. 亜鉛イオン要求性タンパク質が R a f — l、 PKC a、 GEF— H l、 H DAC、 SQSTM1、 ュビキチン一タンパク質リガーゼ E 3、 ュビキチン共役 タンパク質 E 2、 メタロチォネイン、 フォスファターゼ、 S n a i 1、 TRAF 6、 P a x i 1 1 i n、 Z y x i n及び J a d e— 1からなる群より選択される 少なく とも 1種である、 請求項 3 1記載の方法。 32. Zinc ion-requiring protein is R af — l, PKC a, GEF— H l, H DAC, SQSTM1, ubiquitin monoprotein ligase E 3, ubiquitin conjugate protein E 2, metallothionein, phosphatase, S nai 1, TRAF 6, The method according to claim 31, wherein the method is at least one selected from the group consisting of P axi 1 1 in, Z yxin and J ade-1.
33. 亜鉛イオン濃度の制御が亜鉛イオントランスポーターの発現及び Z又は機 能を調節することによって行われるものである、 請求項 26記載の方法。 33. The method of claim 26, wherein the zinc ion concentration is controlled by adjusting the expression and Z or function of the zinc ion transporter.
34. 亜鉛イオントランスポーターが L I Vファミ リ一を含むヒ ト Z I P類、 ヒ ト CD F類からなる群より選択される少なくとも 1種である、 請求項 3 3記載の 方法。 34. The method according to claim 33, wherein the zinc ion transporter is at least one selected from the group consisting of human Z IPs and human CD Fs containing L I V family.
35. 抗原提示細胞が榭状細胞、マクロファージ、 ランゲルハンス細胞、 B細胞、 胸腺上皮細胞、 滑膜細胞、 血管内皮細胞及びケラチノサイ トからなる群より選ば れる少なく とも 1種である、 請求項 26〜34のいずれか 1項に記載の方法。 35. The antigen-presenting cell is at least one selected from the group consisting of rod-shaped cells, macrophages, Langerhans cells, B cells, thymic epithelial cells, synovial cells, vascular endothelial cells, and keratinocytes. The method according to any one of the above.
36. 細胞内の亜鉛イオン濃度を測定することを含む、 抗原提示細胞の機能を制 御し得る化合物のスクリーニング方法。 36. A screening method for a compound capable of controlling the function of antigen-presenting cells, comprising measuring intracellular zinc ion concentration.
37. 抗原提示細胞が樹状細胞、マクロファージ、 ランゲルハンス細胞、 B細胞、 胸腺上皮細胞、 滑膜細胞、 血管内皮細胞及びケラチノサイ トからなる群より選ば れる少なくとも 1種である、 請求項 36記載の方法。 37. The method according to claim 36, wherein the antigen-presenting cell is at least one selected from the group consisting of dendritic cells, macrophages, Langerhans cells, B cells, thymic epithelial cells, synovial cells, vascular endothelial cells, and keratinocytes. .
38. 抗原提示細胞が樹状細胞である、 請求項 3 7記載の方法。 38. The method according to claim 37, wherein the antigen-presenting cell is a dendritic cell.
39. 抗原提示細胞の機能制御剤を製造するための、 細胞內の亜鉛イオン濃度を 制御し得る物質の使用。  39. Use of a substance capable of controlling the zinc ion concentration in the cell wall to produce a function control agent for antigen presenting cells.
40. 免疫システムが関与する疾患の予防 '治療薬を製造するための、 請求項 1 ~ 1 1のいずれか 1項に記載の機能制御剤の使用。 40. Use of a function-controlling agent according to any one of claims 1 to 11 for the manufacture of a prophylactic agent for a disease associated with an immune system.
4 1. 免疫システムが関与する疾患の予防■治療薬を製造するための、 細胞内の 亜鉛ィオン濃度を制御し得る物質の使用。 4 1. Prevention of diseases involving the immune system ■ Use of substances that can control the concentration of intracellular zinc ions to produce therapeutic drugs.
PCT/JP2006/301933 2005-01-31 2006-01-31 Agent for control of function of antigen-presenting cell WO2006080582A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US11/815,071 US20090054316A1 (en) 2005-01-31 2006-01-31 Agent for control of function of antigen-presenting cell
EP06713076A EP1854483A4 (en) 2005-01-31 2006-01-31 Agent for control of function of antigen-presenting cell

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2005-023200 2005-01-31
JP2005023200A JP2006206538A (en) 2005-01-31 2005-01-31 Functional controlling agent for antigen presenting cell

Publications (1)

Publication Number Publication Date
WO2006080582A1 true WO2006080582A1 (en) 2006-08-03

Family

ID=36740586

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2006/301933 WO2006080582A1 (en) 2005-01-31 2006-01-31 Agent for control of function of antigen-presenting cell

Country Status (4)

Country Link
US (1) US20090054316A1 (en)
EP (1) EP1854483A4 (en)
JP (1) JP2006206538A (en)
WO (1) WO2006080582A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008066064A1 (en) * 2006-11-28 2008-06-05 Riken Regulation of the activity of transcriptional factor stat by zinc ion
US9896420B2 (en) 2011-03-10 2018-02-20 The Trustees Of Columbia University In The City Of New York N-quinolin-benzensulfonamides and related compounds for the treatment of cancer, autoimmune disorders and inflammation

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103223165B (en) 2007-08-24 2015-02-11 学校法人庆应义塾 Reducer of immunosuppression by tumor cell and antitumor agent using the same
JP6314201B1 (en) * 2016-11-21 2018-04-18 テラファーマ株式会社 Dendritic cell washing solution, dendritic cell washing method using the same, and method for preparing dendritic cell-containing composition
JP6385607B2 (en) * 2018-02-13 2018-09-05 テラファーマ株式会社 Dendritic cell vaccine

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2003503346A (en) * 1999-06-23 2003-01-28 ジンク セラピューテイクス カナダ インコーポレイテッド Zinc ionophore as an anti-apoptotic agent
WO2003055462A2 (en) * 2001-12-27 2003-07-10 Laboratoires Expanscience Composition comprising at least an alkanolamide for inhibiting langerhans cell migration, and uses thereof

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5409905A (en) * 1981-01-05 1995-04-25 Eby, Iii; George A. Cure for commond cold
US5624675A (en) * 1989-06-06 1997-04-29 Kelly; Patrick D. Genital lubricants containing zinc salts to reduce risk of HIV infection
FR2834216B1 (en) * 2001-12-27 2004-04-30 Pharmascience Lab COSMETIC OR PHARMACEUTICAL COMPOSITION COMPRISING AT LEAST ONE OXAZOLINE FOR INHIBITING LANGERHAN CELL MIGRATION, AND USES THEREOF
US20040180006A1 (en) * 2003-03-12 2004-09-16 Sceusa Nicholas A. Methods and compositions for blocking the calcium cascade

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2003503346A (en) * 1999-06-23 2003-01-28 ジンク セラピューテイクス カナダ インコーポレイテッド Zinc ionophore as an anti-apoptotic agent
WO2003055462A2 (en) * 2001-12-27 2003-07-10 Laboratoires Expanscience Composition comprising at least an alkanolamide for inhibiting langerhans cell migration, and uses thereof

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
CANZONIERO L. M. T. ET AL.: "Membrane-permeant chelators can attenuate Zn2+-induced cortical neuronal death", NEUROPHARMACOLOGY, vol. 45, no. 3, 2003, pages 420 - 428, XP003001969 *
HO L. H. ET AL.: "Labite Zinc and Zinc Transporter ZnT4 in Mast Cell Granules: Role in Regulation of Caspase Activation and NF- kappaB Translocation", JOURNAL OF IMMUNOLOGY, vol. 172, no. 12, 2004, pages 7750 - 7760, XP003001967 *
ITO M. ET AL.: "Anti-allergic and Anti-inflammatory Reaction by Topical Application of Chelating Agents", THE SOCIETY OF COSMETIC CHEMISTS OF JAPAN, vol. 31, no. 3, 1997, pages 322 - 328, XP003001966 *
NOGUEIRA C. W. ET AL.: "2,3-Dimercaptopropanol, 2, 3-dimercaptopropane-1-sulfonic acid, and meso-2, 3-dimercaptosuccinic acid inhibit delta-aminolevulinate dehydratase from human erythrocytes in vitro", ENVIRONMENTAL RESEARCH, vol. 94, no. 3, 2004, pages 254 - 261, XP003001968 *
See also references of EP1854483A4 *
TAYLOR K. M. ET AL.: "Structure-function analysis of HKE4, a member of the new LIV-1 subfamily of zinc transporters", BIOCHEM, J., vol. 377, 2004, pages 131 - 139, XP003004584 *
WEBSTER J. M. ET AL.: "N,N,N',N'-Tetrakis (2-pyridylmethyl)ethylenediamine inhibits ligand binding to certain G protein-coupled receptors", EUROPEAN JOURNAL OF PHARMACOLOGY, vol. 474, no. 1, 2003, pages 1 - 5, XP003001970 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008066064A1 (en) * 2006-11-28 2008-06-05 Riken Regulation of the activity of transcriptional factor stat by zinc ion
US9896420B2 (en) 2011-03-10 2018-02-20 The Trustees Of Columbia University In The City Of New York N-quinolin-benzensulfonamides and related compounds for the treatment of cancer, autoimmune disorders and inflammation

Also Published As

Publication number Publication date
EP1854483A4 (en) 2010-08-04
JP2006206538A (en) 2006-08-10
US20090054316A1 (en) 2009-02-26
EP1854483A1 (en) 2007-11-14

Similar Documents

Publication Publication Date Title
Vogel et al. Cross-talk between aryl hydrocarbon receptor and the inflammatory response: a role for nuclear factor-κB
Quinn et al. The role of Ets2 transcription factor in the induction of microRNA-155 (miR-155) by lipopolysaccharide and its targeting by interleukin-10
Benhamron et al. Regulated IRE1‐dependent decay participates in curtailing immunoglobulin secretion from plasma cells
EP2959918B1 (en) Anti-malignant tumor agent composition
Yang et al. FoxO1 is a regulator of MHC-II expression and anti-tumor effect of tumor-associated macrophages
CN110381963A (en) It is related to the immunotherapy method and composition of tryptophan metabolic pathway regulator
JP2019505498A (en) Inhibition of cytokine-induced SH2 protein in NK cells
Islam et al. Pin1 regulates osteoclast fusion through suppression of the master regulator of cell fusion DC‐STAMP
JP7107839B2 (en) Combination formulations of PKM2 modulators and HMGB1
Correa et al. The NLR-related protein NWD1 is associated with prostate cancer and modulates androgen receptor signaling
Lee et al. Dehydrocostus lactone suppresses osteoclast differentiation by regulating NFATc1 and inhibits osteoclast activation through modulating migration and lysosome function
WO2006080582A1 (en) Agent for control of function of antigen-presenting cell
WO2018023108A1 (en) Trim proteins and galectins cooperate and codirect autophagy and are useful in the treatment of autophagy related diseases
Neff et al. ERK 1/2-and JNKs-dependent synthesis of interleukins 6 and 8 by fibroblast-like synoviocytes stimulated with protein I/II, a modulin from oral streptococci, requires focal adhesion kinase
Zhuo et al. CSTP1 inhibits IL-6 expression through targeting Akt/FoxO3a signaling pathway in bladder cancer cells
CN114929860A (en) Methods and compositions for modulating cellular aging
US20210205300A1 (en) Polycomb inhibitors and uses thereof
JP2021518751A (en) How to adjust Ras ubiquitination
Schultz et al. Gamma secretase dependent release of the CD44 cytoplasmic tail upregulates IFI16 in cd44-/-tumor cells, MEFs and macrophages
Ming-Ming et al. Polyphyllin I promotes cell death via suppressing UPR-mediated CHOP ubiquitination and degradation in non-small cell lung cancer
JPWO2006080581A1 (en) Regulator of degranulation and cytokine production
Saadh et al. Molecular mechanisms of long non-coding RNAs in differentiation of T Helper17 cells
Lin et al. The Heightened Importance of EZH2 in Cancer Immunotherapy
Zhao et al. The unfolded protein response in triple-negative breast cancer
EP3978021A1 (en) Expression regulator of p2x7 receptor

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application
NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2006713076

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 11815071

Country of ref document: US

WWP Wipo information: published in national office

Ref document number: 2006713076

Country of ref document: EP